US9174977B2 - 2-azabicyclo[4.1.0]heptane derivatives as orexin receptor antagonists for the treatment of certain disorders - Google Patents

2-azabicyclo[4.1.0]heptane derivatives as orexin receptor antagonists for the treatment of certain disorders Download PDF

Info

Publication number
US9174977B2
US9174977B2 US14/386,109 US201314386109A US9174977B2 US 9174977 B2 US9174977 B2 US 9174977B2 US 201314386109 A US201314386109 A US 201314386109A US 9174977 B2 US9174977 B2 US 9174977B2
Authority
US
United States
Prior art keywords
methyl
azabicyclo
pyrimidin
mmol
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US14/386,109
Other versions
US20150065523A1 (en
Inventor
Luigi Piero Stasi
Lucio Claudio Rovati
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Rottapharm Biotech SRL
Original Assignee
Rottapharm Biotech SRL
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Rottapharm Biotech SRL filed Critical Rottapharm Biotech SRL
Publication of US20150065523A1 publication Critical patent/US20150065523A1/en
Assigned to ROTTAPHARM BIOTECH S.R.L. reassignment ROTTAPHARM BIOTECH S.R.L. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ROVATI, LUCIO CLAUDIO, STASI, LUIGI PIERO
Application granted granted Critical
Publication of US9174977B2 publication Critical patent/US9174977B2/en
Expired - Fee Related legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/30Drugs for disorders of the nervous system for treating abuse or dependence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Definitions

  • the invention relates to novel 2-azabicyclo[4.1.0]heptane derivatives and their use as pharmaceuticals.
  • the invention also concerns a process for the preparation of those compounds, pharmaceutical compositions containing one or more compounds of formula (I) and their use as dual antagonists of the Orexin 1 and Orexin 2 receptors.
  • Orexin (or hypocretin) signaling is mediated by two receptors and two peptide agonists.
  • the two orexin peptides herein after referred to as orexins, bind to two high affinity receptors, termed Orexin-1 and Orexin-2 receptors.
  • the Orexin-1 receptor is selective in favour of orexin A, while the Orexin-2 receptor binds both orexins with similar affinities.
  • the orexins are cleavage products of the same gene, prepro-orexin.
  • neuronal neurosci the precursor from which orexin is produced
  • neuronal hypothalamus the dorsal hypothalamus and the lateral hypothalamus
  • Orexinergic cells in these nuclei project to many areas of the brain, extending rostrally to the olfactory bulbs and caudally to the spinal cord (van den Pol, A. N. et al., J. Neuroscience., 1999, 19(8), 3171-3182).
  • TMN tuberomammillary nucleus
  • narcolepsy Rodents whose prepro-orexin gene has been knocked out, or whose orexigenic neurons have been lesioned, display altered sleep/wake cycles similar to narcolepsy (Chemelli et al., Cell 1999, 98, 437-451; Hara et al., 2001, supra). Dog models of narcolepsy have been shown to have mutant or nonfunctional orexin-2 receptors (Lin et al., Cell 1999, 98, 365-376). Human narcolepsy appears to be linked to deficient orexin signalling, likely related to immune ablation of orexinergic neurons in the lateral hypothalamus (Mignot et al., Am. J. Hum. Genet.
  • EEG data indicates that orexin-2 may be more important than orexin-1 in the modulation of sleep/wake (P. Malherbe et al., Molecular Pharmacology (2009) 76(3):618-31; C. Dugovic et al., J. Pharmacol. Exp. Ther., 2009, 330(1), 142-151).
  • Disorders of the sleep-wake cycle are therefore likely targets for orexin-2 receptor antagonist therapy. Examples of such disorders include sleep-wake transition disorders, insomnia, restless legs syndrome, jet-lag, disturbed sleep, and sleep disorders secondary to neurological disorders (e.g., manias, depressions, manic depression, schizophrenia, and pain syndromes (e.g., fibromyalgia, neuropathic pain).
  • orexin-2 modulators may be useful to treat various neurological disorders; e.g., agonists or up-regulators to treat catatonia, antagonists or down-regulators to treat Parkinson's disease, Tourette's syndrome, anxiety, delirium and dementias. Recent evidence indicates a role for orexin in the pathogenesis of Alzheimer disease (Kang et al, Science Express, 2009, 1-10).
  • Brain interstitial fluid levels of amyloid-beta were demonstrated to fluctuate diurnally in both humans and rodents with sleep deprivation in rodents leading to significant increases in brain interstitial fluid levels of amyloid-beta.
  • Infusion of a dual orexin antagonist in rodents suppressed interstitial levels of amyloid-beta and abolished the natural diurnal variation of amyloid-beta.
  • the reduction of interstitial fluid amyloid-beta levels is correlated with reduced amyloid plaque formation, a hallmark of Alzheimer's disease, and consequently the regulation of sleep time could potentially inhibit amyloid-beta aggregation and slow the progression of Alzheimer's disease.
  • nicotine J. K. Kane et al., Endocrinology, 2000, 141 (10), 3623-3629; J. K. Kane et al., Neurosci. Lett, 2001, 298(1), 1-4), morphine (D.
  • Orexins and their receptors have been found in both the myenteric and submucosal plexus of the enteric nervous system, where orexins have been shown to increase motility in vitro (Kirchgessner & Liu, Neuron 1999, 24, 941-951) and to stimulate gastric acid secretion in vitro (Takahashi et al., Biochem. Biophys. Res. Comm. 1999, 254, 623-627).
  • Orexin mediated effects on the gut may be driven by a projection via the vagus nerve (van den Pol, 1999, supra), as vagotomy or atropine prevent the effect of an intracerebroventricular injection of orexin on gastric acid secretion (Takahashi et al., 1999, supra).
  • Orexin receptor antagonists or other down-regulators of orexin receptor-mediated systems are therefore potential treatments for ulcers, irritable bowel syndrome, diarrhea and gastroesophageal reflux.
  • Body weight may also be affected by orexin-mediated regulation of appetite and metabolism (T. Sakurai et al., Cell, 1998, 92(4), 573-585; T. Sakurai, Reg. Pept, 1999, 85(1), 25-30).
  • Orexin receptor antagonists therefore are likely to be useful in treatment of overweight or obesity and conditions related to overweight or obesity, such as insulin resistance, type II diabetes, hyperlipidemia, gallstones, angina, hypertension, breathlessness, tachycardia, infertility, sleep apnea, back and joint pain, varicose veins and osteoarthritis.
  • orexin receptor agonists are likely to be useful in treatment of underweight and related conditions such as hypotension, bradycardia, amenorrhea and related infertility, and eating disorders such as anorexia and bulimia.
  • Intracerebroventricularly administered orexins have been shown to increase mean arterial pressure and heart rate in freely moving (awake) animals (Samson et al., Brain Res. 1999, 831, 248-253; Shirasaka et al., Am. J. Physiol. 1999, 277, R1780-R1785) and in urethane-anesthetized animals (Chen et al., Am. J. Physiol. 2000, 278, R692-R697), with similar results.
  • Orexin receptor agonists may therefore be candidates for treatment of hypotension, bradycardia and heart failure related thereto, while orexin receptor antagonists may be useful for treatment of hypertension, tachycardia and other arrhythmias, angina pectoris and acute heart failure.
  • the object of the present invention is to provide 2-azabicyclo[4.1.0]heptane compounds with dual antagonist activity at the Orexin 1 and Orexin 2 receptors.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof:
  • stereochemical isomers enriched in configuration (S) of formula (I) correspond in one embodiment to at least 90% e.e. In another embodiment the isomers correspond to at least 95% e.e. In another embodiment the -isomers correspond to at least 99% e.e.
  • This invention includes in its scope of protection all the possible isomers and racemic mixtures. Wherever should be present further symmetry centres, this invention includes all the possible diastereoisomers and relative mixtures as well.
  • the present invention provides a compound of formula (II), corresponding to a compound of formula (I) in which
  • X is N—H
  • A is a phenyl derivative
  • B is a pyrimidinyl derivative
  • R and R 1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
  • compositions comprising a compound of formula (I) and a pharmaceutically acceptable carrier.
  • the invention concerns a compound of Formula (I) as medicament; in particular it concerns its use for the manufacturing of a medicament for the treatment of pathologies where an antagonist of the OX1/OX2 antagonist is needed, such as the treatment of obesity, sleep disorders, compulsive disorders, drug dependency and schizophrenia.
  • the present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof:
  • 5- or 6-membered heteroaryl ring refers to a monocyclic 5- or 6-membered heterocyclic group containing 1 to 3 heteroatoms and having at least one heteroatom selected from nitrogen, oxygen and sulfur, and containing at least 1 carbon atom.
  • 5 and 6-membered heteroaryl groups include pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, thiazolyl, furyl, thienyl, thiadiazolyl, pyridyl, triazolyl, triazinyl, pyridazinyl, pyrimidinyl and pyrazinyl.
  • C1-C4 alkyl refers to an alkyl group having from one to four carbon atoms, in all isomeric forms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl and tert-butyl.
  • n-C1-C4 alkyl refers to the unbranched alkyls as defined above.
  • C1-C4 alkoxy refers to a straight chain or branched chain alkoxy (or “alkyloxy”) group having from one to four carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy and tert-butoxy.
  • halogen and its abbreviation “halo” refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I). Where the term “halo” is used before another group, it indicates that the group is substituted by one, two or three halogen atoms.
  • haloC 1-4 alkyl refers to groups such as trifluoromethyl, bromoethyl, trifluoropropyl, and other groups derived from C 1-4 alkyl groups as defined above
  • haloC 1-4 alkoxy refers to groups such as trifluoromethoxy, bromoethoxy, trifluoropropoxy, and other groups derived from C 1-4 alkoxy groups as defined above.
  • Any of these groups may be attached to the rest of the molecule at any suitable position.
  • salt refers to any salt of a compound according to the present invention prepared from an inorganic or organic acid or base, quaternary ammonium salts and internally formed salts.
  • Physiologically acceptable salts are particularly suitable for medical applications because of their greater aqueous solubility relative to the parent compounds. Such salts must clearly have a physiologically acceptable anion or cation.
  • physiologically acceptable salts of the compounds of the present invention include acid addition salts formed with inorganic acids such as hydrochloric, hydrobromic, hydroiodic, phosphoric, metaphosphoric, nitric and sulfuric acids, and with organic acids, such as tartaric, acetic, trifluoroacetic, citric, malic, lactic, fumaric, benzoic, formic, propionic, glycolic, gluconic, maleic, succinic, camphorsulfuric, isothionic, mucic, gentisic, isonicotinic, saccharic, glucuronic, furoic, glutamic, ascorbic, anthranilic, salicylic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, pantothenic, stearic, sulfinilic, alginic, galacturonic and arylsulfonic, for example benzenesul, in
  • Pharmaceutical acceptable salts may also be prepared from other salts, including other pharmaceutically acceptable salts, of the compound of formula (I) using conventional methods.
  • prodrugs are also included within the context of this invention.
  • the term “prodrug” means a compound which is converted within the body, e.g. by hydrolysis in the blood, into its active form that has medical effects.
  • Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and in D. Fleisher, S. Ramon and H. Barbra “Improved oral drug delivery: solubility limitations overcome by the use of prodrugs”, Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each of which are incorporated herein by reference.
  • Prodrugs are any covalently bonded carriers that release a compound of structure (I) in vivo when such prodrug is administered to a patient.
  • Prodrugs are generally prepared by modifying functional groups in a way such that the modification is cleaved, either by routine manipulation or in vivo, yielding the parent compound.
  • Prodrugs include, for example, compounds of this invention wherein hydroxy, amine or sulfhydryl groups are bonded to any group that, when administered to a patient, cleaves to form the hydroxy, amine or sulfhydryl groups.
  • prodrugs include (but are not limited to) acetate, formate and benzoate derivatives of alcohol, sulfhydryl and amine functional groups of the compounds of structure (I).
  • esters may be employed, such as methyl esters, ethyl esters, and the like. Esters may be active in their own right and/or be hydrolysable under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those which break down readily in the human body to leave the parent acid or its salt.
  • crystalline forms of the compounds of structure (I) may exist as polymorphs, which are included in the present invention.
  • Suitable protecting groups for use according to the present invention are well known to those skilled in the art and may be used in a conventional manner. See, for example, “Protective groups in organic synthesis” by T. W. Greene and P. G. M. Wuts (John Wiley & sons 1991) or “Protecting Groups” by P. J. Kocienski (Georg Thieme Verlag 1994).
  • suitable amino protecting groups include acyl type protecting groups (e.g.
  • aromatic urethane type protecting groups e.g. benzyloxycarbonyl (Cbz) and substituted Cbz
  • aliphatic urethane protecting groups e.g. 9-fluorenylmethoxycarbonyl (Fmoc), t-butyloxycarbonyl (Boc), isopropyloxycarbonyl, cyclohexyloxycarbonyl
  • alkyl type protecting groups e.g. benzyl, trityl, chlorotrityl.
  • oxygen protecting groups may include for example alky silyl groups, such as trimethylsilyl or tert-butyldimethylsilyl; alkyl ethers such as tetrahydropyranyl or tert-butyl; or esters such as acetate
  • the subject invention also includes isotopically-labelled compounds, which are identical to those recited in formula (I) and following, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulphur, fluorine, iodine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O, 18 O, 31 P, 32 P, 35 S, 18 F, 36 Cl, 123 I and 125 I.
  • Isotopically-labelled compounds of the present invention for example those into which radioactive isotopes such as 3 H, 14 C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3 H, and carbon-14, i.e., 14 C, isotopes are particularly preferred for their ease of preparation and detectability.
  • 11 C and 18 F isotopes are particularly useful in PET (positron emission tomography), and 125 I isotopes are particularly useful in SPECT (single photon emission computerized tomography), all useful in brain imaging.
  • substitution with heavier isotopes such as deuterium, i.e., 2 H can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances.
  • Isotopically labelled compounds of formula (I) and following of this invention can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples below, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
  • Certain groups/substituents included in the present invention may be present as isomers.
  • the present invention includes within its scope all such isomers, including racemates, enantiomers and mixtures thereof.
  • Certain of the substituted heteroaromatic groups included in compounds of formula (I) may exist in one or more tautomeric forms.
  • the present invention includes within its scope all such tautomeric forms, including mixtures.
  • the compounds or salts of the invention should be interpreted as excluding those compounds (if any) which are so chemically unstable, either per se or in water, that they are clearly unsuitable for pharmaceutical use through all administration routes, whether oral, parenteral or otherwise.
  • Such compounds are known to the skilled chemist.
  • Prodrugs or compounds which are stable ex vivo and which are convertible in the mammalian (e.g. human) body to the inventive compounds are however included.
  • the present invention provides a compound of formula (II), corresponding to a compound of formula (I) in which
  • X is N—H
  • A is a phenyl derivative
  • B is a pyrimidinyl derivative
  • R and R 1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
  • the present invention provides a compound of formula (III), corresponding to a compound of formula (I) in which
  • X is N—H
  • A is a thiazolyl derivative
  • B is a phenyl derivative and X is N—H
  • A is a phenyl derivative
  • B is a pyrimidinyl derivative
  • R and R 1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; p is 0 or 1.
  • the present invention provides a compound of formula (IV), corresponding to a compound of formula (I) in which
  • X is O
  • A is a phenyl derivative
  • B is a pyrimidinyl derivative
  • R and R 1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
  • the present invention provides a compound of formula (V), corresponding to a compound of formula (I) in which
  • X is N—H
  • A is a phenyl derivative
  • B is a pyrimidinyl derivative
  • R and R 1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
  • the present invention provides a compound of formula (VI), corresponding to a compound of formula (I) in which
  • X is N—H
  • A is a thiazolyl derivative
  • B is a phenyl derivative
  • R and R 1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; p is 0 or 1.
  • Example compounds of the invention include:
  • a further aspect of this invention concerns a process for the preparation of a compound of formula (I) comprising the following steps represented in the scheme below:
  • step c) two diastereoisomers are formed: they are indicated as “trans” (product Xa) and “cis” (product Xb) relatively to the stereochemistry of carbon represented with (*).
  • step d When the hydrolysis (step d) was conducted at room temperature the “trans” derivative (XIa) was predominantly obtained; when the hydrolysis was performed at higher temperature both “cis” and “trans” diastereoisomers (XIb and XIa respectively) were obtained.
  • Leaving group is as understood by a skilled chemist, i.e. a group which can be displaced by a nucleophile in e.g. a S N 2, S N 1 or S N Ar type reaction, such as an halogen or a reactive residue of a alkyl/aryl sulphonic acid, for example mesylate, tosylate, triflate.
  • the compounds of formula (I) or their pharmaceutically acceptable salts can be used as medicaments, in particular as antagonists of the Orexin 1/Orexin 2 receptors.
  • pharmaceutically acceptable carrier means solvents, carrier agents, diluting agents and the like which are used in the administration of compounds of the invention.
  • compositions can be administered by parenteral, oral, buccal, sublingual, nasal, rectal and topical or transdermal administration.
  • compositions of this invention suitable for the oral administration will be conveniently discrete units such as tablets, capsules, cachet, powders or pellets, or as liquid suspension.
  • the tablets can contain also suitable excipients routinely used in pharmaceutical field such as pre-gelatinised starch, microcrystalline cellulose, sodium glycolate starch, talc, lactose, magnesium stearate, sucrose, stearic acid and mannitol.
  • compositions for parenteral administration conveniently include sterile preparations.
  • Compositions for topical administration may conveniently be formulated as creams, pastes, oils, ointments, emulsions, foams, gels, drops, spray solutions and transdermal patches.
  • compositions can be effected in a manner which will be familiar to any person skilled in the art (see for example Remington, The Science and Practice of Pharmacy, 21 st Edition (2005), Part 5, “Pharmaceutical Manufacturing” [published by Lippincott Williams & Wilkins]) by bringing the described compounds of formula (I) or their pharmaceutically acceptable salts, optionally in combination with other therapeutically valuable substances, into a galenical administration form together with suitable, non-toxic, inert, therapeutically compatible solid or liquid carrier materials and, if desired, usual pharmaceutical adjuvants.
  • the present invention also relates to a method for the prevention or treatment of a disease or disorder mentioned herein comprising administering to a subject a pharmaceutically effective amount of a compound of formula (I).
  • an effective amount of a pharmaceutical composition according to the invention is administered to a subject suffering from or diagnosed as having such disease, disorder or condition.
  • an “effective amount” means an amount or dose sufficient to generally bring about the desired therapeutic or prophylactic benefit in patients in need of such treatment for the designated disease, disorder or condition.
  • Effective amounts or doses of the compounds of the present invention may be ascertained by routine methods such as modelling, dose escalation studies or clinical trials, and by taking into consideration routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the compound, the severity and course of the disease, disorder, or condition, the subject's previous or on-going therapy, the subject's health status and response to drugs, and the judgment of the treating physician.
  • An example of a dose is in the range of from about 0.001 to about 200 mg of compound per kg of subject's body weight per day, preferably about 0.05 to 100 mg/kg/day, or about 1 to 35 mg/kg/day, in single or divided dosage units (e.g., BID, TID, QID).
  • a suitable dosage amount is from about 0.05 to about 7 g/day, or about 0.2 to about 2.5 g/day.
  • the dose may be adjusted for preventative or maintenance treatment.
  • the dosage or the frequency of administration, or both may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained.
  • treatment may cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
  • the compounds according to formula (I) are useful for the prevention or treatment of diseases related to the orexin system.
  • Such diseases related to the orexin system may be selected from the group consisting of all types of sleep disorders, of stress-related syndromes, of addictions (especially psychoactive substance use, abuse, seeking and reinstatement), of cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders, of eating or drinking disorders.
  • such diseases related to the orexin system may be selected from the group consisting of sleep disorders that comprises all types of insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias, restless leg syndrome, sleep apneas, jet-lag syndrome, shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders (notably all types of insomnias, especially primary insomnia).
  • sleep disorders that comprises all types of insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias, restless leg syndrome, sleep apneas, jet-lag syndrome, shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders (notably all types of insomnias, especially primary insomnia).
  • such diseases related to the orexin system may be selected from the group consisting of cognitive dysfunctions that comprise deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
  • such diseases related to the orexin system may be selected from the group consisting of eating disorders that comprise metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa.
  • such diseases related to the orexin system may be selected from the group consisting of all types of addictions (especially psychoactive substance use, abuse, seeking and reinstatement) that comprise all types of psychological or physical addictions and their related tolerance and dependence components.
  • Eating disorders may be defined as comprising metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa.
  • Pathologically modified food intake may result from disturbed appetite (attraction or aversion for food); altered energy balance (intake vs. expenditure); disturbed perception of food quality (high fat or carbohydrates, high palatability); disturbed food availability (unrestricted diet or deprivation) or disrupted water balance.
  • Drinking disorders include polydipsias in psychiatric disorders and all other types of excessive fluid intake.
  • Sleep disorders include all types of parasomnias, insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias; restless leg syndrome; sleep apneas; jet-lag syndrome; shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders.
  • Insomnias are defined as comprising sleep disorders associated with aging; intermittent treatment of chronic insomnia; situational transient insomnia (new environment, noise) or short-term insomnia due to stress; grief; pain or illness. Insomnia also include stress-related syndromes including post-traumatic stress disorders as well as other types and subtypes of anxiety disorders such as generalized anxiety, obsessive compulsive disorder, panic attacks and all types of phobic anxiety and avoidance.
  • Addictions may be defined as addiction to one or more rewarding stimuli, notably to one rewarding stimulus. Such rewarding stimuli may be of either natural or synthetic origin.
  • Psychoactive substance use, abuse, seeking and reinstatement are defined as all types of psychological or physical addictions and their related tolerance and dependence components.
  • Cognitive dysfunctions include deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
  • any characteristics described in this invention for the compounds of formula (I) (whether for the compounds themselves, salts thereof, compositions containing the compounds or salts thereof, uses of the compounds or salts thereof, etc.) apply mutatis mutandis to compounds of formula (II), (III), (IV), (V) and (VI).
  • Reagents used in the following examples were commercially available from various suppliers (for example Sigma-Aldrich, Acros or Apollo scientific) and used without further purifications. Solvents were used in dry form. Reactions in anhydrous environment were run under a positive pressure of dry N 2 .
  • Microwave reactions were run on a Biotage Initiator 2.5 instrument.
  • Mass spectra were run on a Ion Trap Thermo LCQ classic spectrometer, operating in positive ES(+) and negative ES( ⁇ ) ionization mode.
  • UPLC spectra were performed on a Waters Acquity UPLC-SQD instrument using an Acquity UPLC-BEH C18 column (1.7 ⁇ M, 50 ⁇ 2.1 mm).
  • Flash silica gel chromatography was performed on Biotage automatic flash chromatography systems (Sp1 and Isolera systems) using Biotage SNAP HP silica cartridges or Biotage SNAP KP-NH cartridges.
  • DEAD diethylazodicarboxylate
  • DIPEA N,N-diisopropylethylamine
  • Boc terbutyloxycarbonyl
  • DCM dichloromethane
  • TFA trifluoroacetic acid
  • DMF dimethylformamide
  • THF tetrahydrofuran
  • RT room temperature
  • DMAP dimethylamino pyridine
  • AcOEt ethyl acetate.
  • 2-methyl-5-phenylthiazole-4-carboxylic acid may be prepared as in the procedure described in U.S. Pat. No. 3,282,927.
  • the antagonistic activity against human OX1 and OX2 receptors is determined by using CHO e HEK-293 cells transfected with human recombinant OX1 and OX2 receptors respectively, seeded at density of 2 and 3 ⁇ 10 4 cells/well respectively in a 96 fluorimetry well plate.
  • the plate was loaded with the calcium dye (Fluo-4NW/probenecid in HBSS, Hepes 20 mM, pH 7,4; Invitrogen) at 37° C. for 60 min. Afterward the temperature was equilibrated at 22° C. for 15 min and the [Ca2+]i measured directly on the plate by using a fluorescent plate reader (CellLux Perkin Elmer).
  • Invention compounds were dissolved in DMSO, diluted in HBSS (DMSO, 0.3% final) and added to the wells. After 5 min CHO cells were activated with orexin-A, 3 nM while HEK-293 cells were activated with orexin-B, 10 nM.
  • the antagonistic activity has been expressed as pKb (co-logarithm of the apparent dissociation constant calculated by using the modified Cheng Prusoff equation).
  • control specific antagonist response ((measured specific response/control specific agonist response) ⁇ 100) obtained in the presence of the test compounds.
  • the IC 50 values (concentration causing a half-maximal inhibition of the control specific agonist response) were determinated by non-linear regression analysis of the concentration curves generated with mean replicate values using hill equation curve fitting.
  • the IC 50 values are obtained by the arithmetical mean of at least two experiments.

Landscapes

  • Organic Chemistry (AREA)
  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Psychiatry (AREA)
  • Child & Adolescent Psychology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Addiction (AREA)
  • Anesthesiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention provides compounds of formula (I) including stereoisomers or a racemate or a mixture or a pharmaceutically acceptable salt thereof: wherein X is NH, or O; Q is 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; A is phenyl or a 5-6 heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; B may assume different meanings from A and is phenyl or a 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; and processes for their preparation, pharmaceutical compositions containing them and their use as dual antagonists of the Orexin 1 and Orexin 2 receptors.
Figure US09174977-20151103-C00001

Description

This application is a national stage application of International Application No. PCT/EP2013/055548, filed Mar. 18, 2013, which claims the benefit of Italian Patent Application No. MI2012A000424, filed Mar. 19, 2012, each of which is incorporated herein by reference in its entirety.
FIELD OF THE INVENTION
The invention relates to novel 2-azabicyclo[4.1.0]heptane derivatives and their use as pharmaceuticals. The invention also concerns a process for the preparation of those compounds, pharmaceutical compositions containing one or more compounds of formula (I) and their use as dual antagonists of the Orexin 1 and Orexin 2 receptors.
BACKGROUND OF THE INVENTION
Orexin (or hypocretin) signaling is mediated by two receptors and two peptide agonists. The two orexin peptides (orexin A and orexin B) herein after referred to as orexins, bind to two high affinity receptors, termed Orexin-1 and Orexin-2 receptors. The Orexin-1 receptor is selective in favour of orexin A, while the Orexin-2 receptor binds both orexins with similar affinities. The orexins, are cleavage products of the same gene, prepro-orexin. In the central nervous system neurons expressing prepro-orexin, the precursor from which orexin is produced, are found in the perifornical nucleus, the dorsal hypothalamus and the lateral hypothalamus (C. Peyron et al., J. Neurosci., 1998, 18(23), 9996-10015). Orexinergic cells in these nuclei project to many areas of the brain, extending rostrally to the olfactory bulbs and caudally to the spinal cord (van den Pol, A. N. et al., J. Neuroscience., 1999, 19(8), 3171-3182).
The broad CNS distribution of orexin projections and neurons expressing orexin receptors is suggestive of orexin involvement in a number of physiological functions including; feeding, drinking, arousal, stress, reward, metabolism and reproduction (T. Sakurai, Nature Reviews Neuroscience, 2007, 8(3), 171-181). The targeted necrosis of cells expressing prepro-orexin suggests the most physiologically important roles of the orexins are likely to be effects on arousal, feeding and metabolism (J. Hara et al., Neuron, 2001, 30, 345-354). A prominent orexin neuronal projection via the vagus nerve probably mediates central orexin effects on cardiac parameters (W. K. Samson et al., Brain Res., 1999, 831, 248-253; T. Shirasaka et al., Am. J. Physiol., 1999, 277, R1780-R1785; C.-T. Chen et al., Am. J. Physiol., 2000, 278, R692-R697), gastric acid secretion and gastric motility (A. L. Kirchgessner and M.-T. Liu, Neuron, 1999, 24, 941-951; N. Takahashi et al., Biochem. Biophys. Res. Commun., 1999, 254, 623-627).
Several lines of evidence indicate that the orexin system is an important modulator of arousal. Rodents administered orexins intracerebroventricularly spend more time awake (Piper et al., J. Neurosci. 2000, 12, 726-730). Orexin mediated effects on arousal have been linked to orexin neuronal projections to histaminergic neurons in the tuberomammillary nucleus (TMN) (Yamanaka et al., Biochem. Biophys. Res. Comm. 2002, 290, 1237-1245). TMN neurons express the orexin-2 receptor primarily, and the orexin-1 receptor to a lesser extent. Rodents whose prepro-orexin gene has been knocked out, or whose orexigenic neurons have been lesioned, display altered sleep/wake cycles similar to narcolepsy (Chemelli et al., Cell 1999, 98, 437-451; Hara et al., 2001, supra). Dog models of narcolepsy have been shown to have mutant or nonfunctional orexin-2 receptors (Lin et al., Cell 1999, 98, 365-376). Human narcolepsy appears to be linked to deficient orexin signalling, likely related to immune ablation of orexinergic neurons in the lateral hypothalamus (Mignot et al., Am. J. Hum. Genet. 2001, 68: 686-699; Minot & Thorsby, New England J. Med. 2001, 344, 692), or, in rare cases, to mutations in the orexin-2 gene (Peyron et al., Nature Med. 2000, 6, 991-997). The disclosure that rats, dogs and humans treated with the dual orexin-1/2 receptor antagonist, ACT-078573 (Brisbare-Roch et al., Nature Medicine, 2007, 13, 150-155) exhibited decreased alertness together with characteristic clinical and EEG (electroencephalographic) signs of sleep provides evidence to support a role for the orexin system in the regulation of arousal, sleep and wake states. EEG data indicates that orexin-2 may be more important than orexin-1 in the modulation of sleep/wake (P. Malherbe et al., Molecular Pharmacology (2009) 76(3):618-31; C. Dugovic et al., J. Pharmacol. Exp. Ther., 2009, 330(1), 142-151). Disorders of the sleep-wake cycle are therefore likely targets for orexin-2 receptor antagonist therapy. Examples of such disorders include sleep-wake transition disorders, insomnia, restless legs syndrome, jet-lag, disturbed sleep, and sleep disorders secondary to neurological disorders (e.g., manias, depressions, manic depression, schizophrenia, and pain syndromes (e.g., fibromyalgia, neuropathic pain). The orexin system also interacts with brain dopamine systems. Intracerebroventricular injections of orexins in mice increase locomotor activity, grooming and stereotypy; these behavioural effects are reversed by administration of D2 dopamine receptor antagonists (Nakamura et al., Brain Research, 873(1), 181-7). Therefore, orexin-2 modulators may be useful to treat various neurological disorders; e.g., agonists or up-regulators to treat catatonia, antagonists or down-regulators to treat Parkinson's disease, Tourette's syndrome, anxiety, delirium and dementias. Recent evidence indicates a role for orexin in the pathogenesis of Alzheimer disease (Kang et al, Science Express, 2009, 1-10). Brain interstitial fluid levels of amyloid-beta were demonstrated to fluctuate diurnally in both humans and rodents with sleep deprivation in rodents leading to significant increases in brain interstitial fluid levels of amyloid-beta. Infusion of a dual orexin antagonist in rodents suppressed interstitial levels of amyloid-beta and abolished the natural diurnal variation of amyloid-beta. The reduction of interstitial fluid amyloid-beta levels is correlated with reduced amyloid plaque formation, a hallmark of Alzheimer's disease, and consequently the regulation of sleep time could potentially inhibit amyloid-beta aggregation and slow the progression of Alzheimer's disease. Orexin neurons project to many regions of the brain associated with reward function (T. Sakurai, supra) and research, focusing on animal models of drug intake, reward, and reinstatement, has expanded the link between the orexin system and addiction. A comprehensive set of data suggest that drugs of abuse activate the orexin system, which in turn enhances drug reward or drug seeking (G. Aston-Jones et al., Neuropharmacology, 2009, 56 (Suppl 1) 1 12-121. Thus interactions between nicotine (J. K. Kane et al., Endocrinology, 2000, 141 (10), 3623-3629; J. K. Kane et al., Neurosci. Lett, 2001, 298(1), 1-4), morphine (D. Georgescu, et al., J. Neurosci., 2003, 23(8), 3106-3111) and amphetamine (C. J. Winrow et al., Neuropharmacology, 2010, 58(1), 185-94) and the orexin system have been demonstrated. Additional studies from a number of laboratories have demonstrated an important relationship between the Orexin system and ethanol consumption. As examples, ethanol consumption in an alcohol-preferring strain of rat was shown to up regulate Orexin mRNA in the lateral hypothalamus and that an Orexin-1 receptor antagonist reduced operant responding for ethanol (Lawrence, et. al., Br. J. Pharmacol., 2006, 148, 752-759). Treatment with an orexin-1 antagonist has also been shown to decrease operant responding for ethanol (Richards, et. al., Psychopharmacology, 2008, 199 (1), 109-117). Other studies have demonstrated increased Fos activation of orexin neurons following contextual reinstatement to ethanol seeking (Dayas, et. al., Biol. Psychiatry, 2008, 63 (2), 152-157 and Hamlin, et. al., Neuroscience, 2007, 146, 525-536). Studies have also shown increased ethanol consumption following Orexin infusion into the paraventricular nucleus of the hypothalamus or in the lateral hypothalamus (Schneider, et. al., Alcohol. Clin. Exp. Res., 2007, 37(11), 1858-1865). These studies provide evidence that modulation of the Orexin system effects alcohol preference and therefore Orexin receptor antagonists are likely to be useful for the treatment of alcoholism.
Orexins and their receptors have been found in both the myenteric and submucosal plexus of the enteric nervous system, where orexins have been shown to increase motility in vitro (Kirchgessner & Liu, Neuron 1999, 24, 941-951) and to stimulate gastric acid secretion in vitro (Takahashi et al., Biochem. Biophys. Res. Comm. 1999, 254, 623-627). Orexin mediated effects on the gut may be driven by a projection via the vagus nerve (van den Pol, 1999, supra), as vagotomy or atropine prevent the effect of an intracerebroventricular injection of orexin on gastric acid secretion (Takahashi et al., 1999, supra). Orexin receptor antagonists or other down-regulators of orexin receptor-mediated systems are therefore potential treatments for ulcers, irritable bowel syndrome, diarrhea and gastroesophageal reflux. Body weight may also be affected by orexin-mediated regulation of appetite and metabolism (T. Sakurai et al., Cell, 1998, 92(4), 573-585; T. Sakurai, Reg. Pept, 1999, 85(1), 25-30). Some effects of orexin on metabolism and appetite may be mediated in the gut, where, as mentioned, orexins alter gastric motility and gastric acid secretion. Orexin receptor antagonists therefore are likely to be useful in treatment of overweight or obesity and conditions related to overweight or obesity, such as insulin resistance, type II diabetes, hyperlipidemia, gallstones, angina, hypertension, breathlessness, tachycardia, infertility, sleep apnea, back and joint pain, varicose veins and osteoarthritis. Conversely, orexin receptor agonists are likely to be useful in treatment of underweight and related conditions such as hypotension, bradycardia, amenorrhea and related infertility, and eating disorders such as anorexia and bulimia. Intracerebroventricularly administered orexins have been shown to increase mean arterial pressure and heart rate in freely moving (awake) animals (Samson et al., Brain Res. 1999, 831, 248-253; Shirasaka et al., Am. J. Physiol. 1999, 277, R1780-R1785) and in urethane-anesthetized animals (Chen et al., Am. J. Physiol. 2000, 278, R692-R697), with similar results.
Orexin receptor agonists may therefore be candidates for treatment of hypotension, bradycardia and heart failure related thereto, while orexin receptor antagonists may be useful for treatment of hypertension, tachycardia and other arrhythmias, angina pectoris and acute heart failure.
From the foregoing discussion, it can be seen that the identification of orexin receptor antagonists, in one embodiment modulators of the orexin-2 receptor, will be of great advantage in the development of therapeutic agents for the treatment of a wide variety of disorders that are mediated through these receptor systems.
Certain orexin antagonists are disclosed in PCT patent applications: WO2010/0480116, WO2010/051238, WO2006/127550, WO2010/060470, WO2010/060471, WO2003/051368, WO2011/076747 and WO2009/016564. There remains a need, however, for potent orexin dual receptor antagonists with desirable pharmaceutical properties
The object of the present invention is to provide 2-azabicyclo[4.1.0]heptane compounds with dual antagonist activity at the Orexin 1 and Orexin 2 receptors.
SUMMARY OF THE INVENTION
The present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure US09174977-20151103-C00002

wherein
    • X is NH, or O;
    • Q is 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN;
    • A is phenyl or a 5-6 heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN;
    • B may assume different meanings from A and is phenyl or a 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN.
Compounds of formula (I) are provided as (S) enantiomers at the chiral carbon represented with an asterisk (*). It is intended in the context of the present invention that stereochemical isomers enriched in configuration (S) of formula (I) correspond in one embodiment to at least 90% e.e. In another embodiment the isomers correspond to at least 95% e.e. In another embodiment the -isomers correspond to at least 99% e.e.
This invention includes in its scope of protection all the possible isomers and racemic mixtures. Wherever should be present further symmetry centres, this invention includes all the possible diastereoisomers and relative mixtures as well.
In a first embodiment, the present invention provides a compound of formula (II), corresponding to a compound of formula (I) in which
Figure US09174977-20151103-C00003

X is N—H, A is a phenyl derivative, B is a pyrimidinyl derivative and R and R1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
In another aspect the invention concerns pharmaceutical compositions comprising a compound of formula (I) and a pharmaceutically acceptable carrier.
In another aspect the invention concerns a compound of Formula (I) as medicament; in particular it concerns its use for the manufacturing of a medicament for the treatment of pathologies where an antagonist of the OX1/OX2 antagonist is needed, such as the treatment of obesity, sleep disorders, compulsive disorders, drug dependency and schizophrenia.
DETAILED DESCRIPTION OF THE INVENTION
The present invention thus provides a compound of formula (I) or a pharmaceutically acceptable salt thereof:
Figure US09174977-20151103-C00004

wherein
    • X is NH, or O;
    • Q is 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN;
    • A is phenyl or a 5-6 heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN;
    • B may assume different meanings from A and is phenyl or a 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN.
The term “5- or 6-membered heteroaryl ring” refers to a monocyclic 5- or 6-membered heterocyclic group containing 1 to 3 heteroatoms and having at least one heteroatom selected from nitrogen, oxygen and sulfur, and containing at least 1 carbon atom. Examples of 5 and 6-membered heteroaryl groups include pyrrolyl, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, isothiazolyl, thiazolyl, furyl, thienyl, thiadiazolyl, pyridyl, triazolyl, triazinyl, pyridazinyl, pyrimidinyl and pyrazinyl.
The term “C1-C4 alkyl” refers to an alkyl group having from one to four carbon atoms, in all isomeric forms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl and tert-butyl. The term “n-C1-C4 alkyl” refers to the unbranched alkyls as defined above.
The term “C1-C4 alkoxy” refers to a straight chain or branched chain alkoxy (or “alkyloxy”) group having from one to four carbon atoms, such as methoxy, ethoxy, propoxy, isopropoxy, butoxy, isobutoxy, sec-butoxy and tert-butoxy.
The term “halogen” and its abbreviation “halo” refers to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I). Where the term “halo” is used before another group, it indicates that the group is substituted by one, two or three halogen atoms. For example, “haloC1-4alkyl” refers to groups such as trifluoromethyl, bromoethyl, trifluoropropyl, and other groups derived from C1-4alkyl groups as defined above; and the term “haloC1-4alkoxy” refers to groups such as trifluoromethoxy, bromoethoxy, trifluoropropoxy, and other groups derived from C1-4alkoxy groups as defined above.
Any of these groups may be attached to the rest of the molecule at any suitable position.
As used herein, the term “salt” refers to any salt of a compound according to the present invention prepared from an inorganic or organic acid or base, quaternary ammonium salts and internally formed salts. Physiologically acceptable salts are particularly suitable for medical applications because of their greater aqueous solubility relative to the parent compounds. Such salts must clearly have a physiologically acceptable anion or cation. Suitably physiologically acceptable salts of the compounds of the present invention include acid addition salts formed with inorganic acids such as hydrochloric, hydrobromic, hydroiodic, phosphoric, metaphosphoric, nitric and sulfuric acids, and with organic acids, such as tartaric, acetic, trifluoroacetic, citric, malic, lactic, fumaric, benzoic, formic, propionic, glycolic, gluconic, maleic, succinic, camphorsulfuric, isothionic, mucic, gentisic, isonicotinic, saccharic, glucuronic, furoic, glutamic, ascorbic, anthranilic, salicylic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, pantothenic, stearic, sulfinilic, alginic, galacturonic and arylsulfonic, for example benzenesulfonic and p-toluenesulfonic, acids; base addition salts formed with alkali metals and alkaline earth metals and organic bases such as N,N-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumaine (N-methylglucamine), lysine and procaine; and internally formed salts. Salts having a non-physiologically acceptable anion or cation are within the scope of the invention as useful intermediates for the preparation of physiologically acceptable salts and/or for use in non-therapeutic, for example, in vitro, situations.
Pharmaceutical acceptable salts may also be prepared from other salts, including other pharmaceutically acceptable salts, of the compound of formula (I) using conventional methods.
Those skilled in the art of organic chemistry will appreciate that many organic compounds can form complexes with solvents in which they are reacted or from which they are precipitated or crystallized. These complexes are known as “solvates”. For example, a complex with water is known as a “hydrate”. Solvates of the compound of the invention are within the scope of the invention. The compounds of formula (I) may readily be isolated in association with solvent molecules by crystallisation or evaporation of an appropriate solvent to give the corresponding solvates.
In addition, prodrugs are also included within the context of this invention. As used herein, the term “prodrug” means a compound which is converted within the body, e.g. by hydrolysis in the blood, into its active form that has medical effects. Pharmaceutically acceptable prodrugs are described in T. Higuchi and V. Stella, Prodrugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, and in D. Fleisher, S. Ramon and H. Barbra “Improved oral drug delivery: solubility limitations overcome by the use of prodrugs”, Advanced Drug Delivery Reviews (1996) 19(2) 115-130, each of which are incorporated herein by reference.
Prodrugs are any covalently bonded carriers that release a compound of structure (I) in vivo when such prodrug is administered to a patient. Prodrugs are generally prepared by modifying functional groups in a way such that the modification is cleaved, either by routine manipulation or in vivo, yielding the parent compound. Prodrugs include, for example, compounds of this invention wherein hydroxy, amine or sulfhydryl groups are bonded to any group that, when administered to a patient, cleaves to form the hydroxy, amine or sulfhydryl groups. Thus, representative examples of prodrugs include (but are not limited to) acetate, formate and benzoate derivatives of alcohol, sulfhydryl and amine functional groups of the compounds of structure (I). Further, in the case of a carboxylic acid (—COOH), esters may be employed, such as methyl esters, ethyl esters, and the like. Esters may be active in their own right and/or be hydrolysable under in vivo conditions in the human body. Suitable pharmaceutically acceptable in vivo hydrolysable ester groups include those which break down readily in the human body to leave the parent acid or its salt.
Furthermore, some of the crystalline forms of the compounds of structure (I) may exist as polymorphs, which are included in the present invention.
Hereinafter, compounds of formula (I) and their pharmaceutically acceptable salts, and solvates defined in any aspect of the invention (except intermediate compounds in chemical processes) are referred to as “compounds of the invention”.
Those skilled in the art will appreciate that in the preparation of the compound of the invention or a solvate thereof it may be necessary and/or desirable to protect one or more sensitive groups in the molecule to prevent undesirable side reactions. Suitable protecting groups for use according to the present invention are well known to those skilled in the art and may be used in a conventional manner. See, for example, “Protective groups in organic synthesis” by T. W. Greene and P. G. M. Wuts (John Wiley & sons 1991) or “Protecting Groups” by P. J. Kocienski (Georg Thieme Verlag 1994). Examples of suitable amino protecting groups include acyl type protecting groups (e.g. formyl, trifluoroacetyl, acetyl), aromatic urethane type protecting groups (e.g. benzyloxycarbonyl (Cbz) and substituted Cbz), aliphatic urethane protecting groups (e.g. 9-fluorenylmethoxycarbonyl (Fmoc), t-butyloxycarbonyl (Boc), isopropyloxycarbonyl, cyclohexyloxycarbonyl) and alkyl type protecting groups (e.g. benzyl, trityl, chlorotrityl). Examples of suitable oxygen protecting groups may include for example alky silyl groups, such as trimethylsilyl or tert-butyldimethylsilyl; alkyl ethers such as tetrahydropyranyl or tert-butyl; or esters such as acetate
When a specific enantiomer of a compound of general formula (I) is required, this may be obtained for example by resolution of a corresponding enantiomeric mixture of a compound of formula (I) using conventional methods. Thus the required enantiomer may be obtained from the racemic compound of formula (I) by use of chiral HPLC procedure.
The subject invention also includes isotopically-labelled compounds, which are identical to those recited in formula (I) and following, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention and pharmaceutically acceptable salts thereof include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulphur, fluorine, iodine, and chlorine, such as 2H, 3H, 11C, 13C, 14C, 15N, 17O, 18O, 31P, 32P, 35S, 18F, 36Cl, 123I and 125I.
Compounds of the present invention and pharmaceutically acceptable salts of said compounds that contain the aforementioned isotopes and/or other isotopes of other atoms are within the scope of the present invention. Isotopically-labelled compounds of the present invention, for example those into which radioactive isotopes such as 3H, 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. 11C and 18F isotopes are particularly useful in PET (positron emission tomography), and 125I isotopes are particularly useful in SPECT (single photon emission computerized tomography), all useful in brain imaging. Further, substitution with heavier isotopes such as deuterium, i.e., 2H, can afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements and, hence, may be preferred in some circumstances. Isotopically labelled compounds of formula (I) and following of this invention can generally be prepared by carrying out the procedures disclosed in the Schemes and/or in the Examples below, by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
Certain groups/substituents included in the present invention may be present as isomers. The present invention includes within its scope all such isomers, including racemates, enantiomers and mixtures thereof. Certain of the substituted heteroaromatic groups included in compounds of formula (I) may exist in one or more tautomeric forms. The present invention includes within its scope all such tautomeric forms, including mixtures.
In general, the compounds or salts of the invention should be interpreted as excluding those compounds (if any) which are so chemically unstable, either per se or in water, that they are clearly unsuitable for pharmaceutical use through all administration routes, whether oral, parenteral or otherwise. Such compounds are known to the skilled chemist. Prodrugs or compounds which are stable ex vivo and which are convertible in the mammalian (e.g. human) body to the inventive compounds are however included.
In a first embodiment, the present invention provides a compound of formula (II), corresponding to a compound of formula (I) in which
Figure US09174977-20151103-C00005

X is N—H, A is a phenyl derivative, B is a pyrimidinyl derivative and R and R1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
In a second embodiment, the present invention provides a compound of formula (III), corresponding to a compound of formula (I) in which
Figure US09174977-20151103-C00006

X is N—H, A is a thiazolyl derivative, B is a phenyl derivative and X is N—H, A is a phenyl derivative, B is a pyrimidinyl derivative and R and R1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; p is 0 or 1.
In a third embodiment, the present invention provides a compound of formula (IV), corresponding to a compound of formula (I) in which
Figure US09174977-20151103-C00007

X is O, A is a phenyl derivative, B is a pyrimidinyl derivative and R and R1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
In a fourth embodiment, the present invention provides a compound of formula (V), corresponding to a compound of formula (I) in which
Figure US09174977-20151103-C00008

X is N—H, A is a phenyl derivative, B is a pyrimidinyl derivative and R and R1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; n is 1, 2, or 3.
In a fifth embodiment, the present invention provides a compound of formula (VI), corresponding to a compound of formula (I) in which
Figure US09174977-20151103-C00009

X is N—H, A is a thiazolyl derivative, B is a phenyl derivative and R and R1 are independently selected from a group consisting of: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, CN; m is 0, 1, 2, 3 or 4; p is 0 or 1.
Example compounds of the invention include:
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino-) methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • 4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
  • ((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-(pyrimidin-2-yl)phenyl)methanone;
  • (5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • 4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
  • 3-((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)-4-(pyrimidin-2-yl)benzonitrile;
  • 4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
  • (5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (2-methyl-5-phenylthiazol-4-yl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • ((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(2-methyl-5-phenylthiazol-4-yl)methanone;
  • (2-methyl-5-phenylthiazol-4-yl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • 3-((1R,3S,6S)-3-(((5-chloropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)-4-(pyrimidin-2-yl)benzonitrile;
  • 3-((1R,3S,6S)-3-(((5-fluoropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)-4-(pyrimidin-2-yl)benzonitrile;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-fluoropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-chloropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • ((1R,3S,6S)-3-(((5-chloropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-(pyrimidin-2-yl)phenyl)methanone;
  • (5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)oxy)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • 4-(pyrimidin-2-yl)-3-((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • ((1S,3S,6R)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-(pyrimidin-2-yl)phenyl)methanone;
  • (5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)-methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
  • (2-methyl-5-phenylthiazol-4-yl)((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone; or their pharmaceutically acceptable salts thereof.
A further aspect of this invention concerns a process for the preparation of a compound of formula (I) comprising the following steps represented in the scheme below:
Figure US09174977-20151103-C00010
    • Step a) means introducing a protecting group, such as BOC, to obtain a compound of formula (VIII);
    • Step b) means reducing with suitable reducing agent, such as LiEt3BH, to obtain a compound of formula (IX)
    • Step c) means reacting a compound of formula (IX) with suitable reagents, such as CH2I2 and Et2Zn, to obtain a compound of formula (X);
    • Step d) means hydrolysis of a compound of formula (X) to obtain a compound of formula (XI);
    • Step e) means reducing with an appropriate reagent, such as BH3, to obtain a compound of formula (XII);
    • Step f) means converting the alcohol with an amine by using a precursor, such as phtalimide, under Mitsunobu conditions to obtain a compound of formula (XIII);
    • Step g) means deprotection of the phtalimide by using a suitable reagent, such as hydrazine, to obtain a compound of formula (XIV);
    • Step h) means adding a compound of formula R—X, where R is defined as above and X is a leaving group, to the compounds of formula (XIV) or (XII) to obtain compounds (XV) and (XVIII) respectively;
    • Step i) means cleaving the protecting group (PG), such as the BOC group from the compounds of formula (XV) and (XVIII) to obtain compounds of Formula (XVI) and (XIX) respectively;
    • Step I) means reacting a compound of Formula (XVI) and (XIX) with RCOOH or a reactive derivative thereof (such as anhydride or acyl chloride) in the presence of coupling reagents in the presence of a base, where P is defined as above.
In the commercially available compound of formula (VII), the absolute stereochemistry of the carbon represented with (*) is (S). As a consequence, the stereochemistry of products of formula (I) to (VI) has been reasonably assigned on the assumption that the absolute configuration at this centre is retained.
During step c) two diastereoisomers are formed: they are indicated as “trans” (product Xa) and “cis” (product Xb) relatively to the stereochemistry of carbon represented with (*).
Figure US09174977-20151103-C00011
When the hydrolysis (step d) was conducted at room temperature the “trans” derivative (XIa) was predominantly obtained; when the hydrolysis was performed at higher temperature both “cis” and “trans” diastereoisomers (XIb and XIa respectively) were obtained.
Figure US09174977-20151103-C00012
“Leaving group” is as understood by a skilled chemist, i.e. a group which can be displaced by a nucleophile in e.g. a SN2, SN1 or SNAr type reaction, such as an halogen or a reactive residue of a alkyl/aryl sulphonic acid, for example mesylate, tosylate, triflate.
The compounds of formula (I) or their pharmaceutically acceptable salts can be used as medicaments, in particular as antagonists of the Orexin 1/Orexin 2 receptors.
They could be used in combination with a pharmaceutically acceptable carrier and, optionally, with suitable excipients, to obtain pharmaceutical compositions. The term “pharmaceutically acceptable carrier” means solvents, carrier agents, diluting agents and the like which are used in the administration of compounds of the invention.
Such pharmaceutical compositions can be administered by parenteral, oral, buccal, sublingual, nasal, rectal and topical or transdermal administration.
Compositions of this invention suitable for the oral administration will be conveniently discrete units such as tablets, capsules, cachet, powders or pellets, or as liquid suspension. The tablets can contain also suitable excipients routinely used in pharmaceutical field such as pre-gelatinised starch, microcrystalline cellulose, sodium glycolate starch, talc, lactose, magnesium stearate, sucrose, stearic acid and mannitol.
Compositions for parenteral administration conveniently include sterile preparations. Compositions for topical administration may conveniently be formulated as creams, pastes, oils, ointments, emulsions, foams, gels, drops, spray solutions and transdermal patches.
The production of the pharmaceutical compositions can be effected in a manner which will be familiar to any person skilled in the art (see for example Remington, The Science and Practice of Pharmacy, 21 st Edition (2005), Part 5, “Pharmaceutical Manufacturing” [published by Lippincott Williams & Wilkins]) by bringing the described compounds of formula (I) or their pharmaceutically acceptable salts, optionally in combination with other therapeutically valuable substances, into a galenical administration form together with suitable, non-toxic, inert, therapeutically compatible solid or liquid carrier materials and, if desired, usual pharmaceutical adjuvants.
The present invention also relates to a method for the prevention or treatment of a disease or disorder mentioned herein comprising administering to a subject a pharmaceutically effective amount of a compound of formula (I).
In treatment methods according to the invention, an effective amount of a pharmaceutical composition according to the invention is administered to a subject suffering from or diagnosed as having such disease, disorder or condition.
An “effective amount” means an amount or dose sufficient to generally bring about the desired therapeutic or prophylactic benefit in patients in need of such treatment for the designated disease, disorder or condition. Effective amounts or doses of the compounds of the present invention may be ascertained by routine methods such as modelling, dose escalation studies or clinical trials, and by taking into consideration routine factors, e.g., the mode or route of administration or drug delivery, the pharmacokinetics of the compound, the severity and course of the disease, disorder, or condition, the subject's previous or on-going therapy, the subject's health status and response to drugs, and the judgment of the treating physician. An example of a dose is in the range of from about 0.001 to about 200 mg of compound per kg of subject's body weight per day, preferably about 0.05 to 100 mg/kg/day, or about 1 to 35 mg/kg/day, in single or divided dosage units (e.g., BID, TID, QID). For a 70-kg human, an illustrative range for a suitable dosage amount is from about 0.05 to about 7 g/day, or about 0.2 to about 2.5 g/day.
Once improvement of the patient's disease, disorder, or condition has occurred, the dose may be adjusted for preventative or maintenance treatment. For example, the dosage or the frequency of administration, or both, may be reduced as a function of the symptoms, to a level at which the desired therapeutic or prophylactic effect is maintained. Of course, if symptoms have been alleviated to an appropriate level, treatment may cease. Patients may, however, require intermittent treatment on a long-term basis upon any recurrence of symptoms.
For avoidance of any doubt, if compounds are described as useful for the prevention or treatment of certain diseases, such compounds are likewise suitable for use in the preparation of a medicament for the prevention or treatment of said diseases.
The compounds according to formula (I) are useful for the prevention or treatment of diseases related to the orexin system.
Such diseases related to the orexin system may be selected from the group consisting of all types of sleep disorders, of stress-related syndromes, of addictions (especially psychoactive substance use, abuse, seeking and reinstatement), of cognitive dysfunctions in the healthy population and in psychiatric and neurologic disorders, of eating or drinking disorders.
In a sub-embodiment, such diseases related to the orexin system may be selected from the group consisting of sleep disorders that comprises all types of insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias, restless leg syndrome, sleep apneas, jet-lag syndrome, shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders (notably all types of insomnias, especially primary insomnia).
In another sub-embodiment, such diseases related to the orexin system may be selected from the group consisting of cognitive dysfunctions that comprise deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
In another sub-embodiment, such diseases related to the orexin system may be selected from the group consisting of eating disorders that comprise metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa.
In another sub-embodiment, such diseases related to the orexin system may be selected from the group consisting of all types of addictions (especially psychoactive substance use, abuse, seeking and reinstatement) that comprise all types of psychological or physical addictions and their related tolerance and dependence components.
Eating disorders may be defined as comprising metabolic dysfunction; dysregulated appetite control; compulsive obesities; emeto-bulimia or anorexia nervosa.
Pathologically modified food intake may result from disturbed appetite (attraction or aversion for food); altered energy balance (intake vs. expenditure); disturbed perception of food quality (high fat or carbohydrates, high palatability); disturbed food availability (unrestricted diet or deprivation) or disrupted water balance. Drinking disorders include polydipsias in psychiatric disorders and all other types of excessive fluid intake.
Sleep disorders include all types of parasomnias, insomnias, narcolepsy and other disorders of excessive sleepiness, sleep-related dystonias; restless leg syndrome; sleep apneas; jet-lag syndrome; shift-work syndrome, delayed or advanced sleep phase syndrome or insomnias related to psychiatric disorders.
Insomnias are defined as comprising sleep disorders associated with aging; intermittent treatment of chronic insomnia; situational transient insomnia (new environment, noise) or short-term insomnia due to stress; grief; pain or illness. Insomnia also include stress-related syndromes including post-traumatic stress disorders as well as other types and subtypes of anxiety disorders such as generalized anxiety, obsessive compulsive disorder, panic attacks and all types of phobic anxiety and avoidance.
Addictions may be defined as addiction to one or more rewarding stimuli, notably to one rewarding stimulus. Such rewarding stimuli may be of either natural or synthetic origin. Psychoactive substance use, abuse, seeking and reinstatement are defined as all types of psychological or physical addictions and their related tolerance and dependence components.
Cognitive dysfunctions include deficits in all types of attention, learning and memory functions occurring transiently or chronically in the normal, healthy, young, adult or aging population, and also occurring transiently or chronically in psychiatric, neurologic, cardiovascular and immune disorders.
Besides, any characteristics described in this invention for the compounds of formula (I) (whether for the compounds themselves, salts thereof, compositions containing the compounds or salts thereof, uses of the compounds or salts thereof, etc.) apply mutatis mutandis to compounds of formula (II), (III), (IV), (V) and (VI).
EXPERIMENTAL SECTION
The invention will be now detailed by means of the following examples relating to the preparation of some invention compounds and to the evaluation of their activity against OX1 receptor and OX2 receptor.
In the procedure that follows, after the starting materials, reference to a description is typically provided. The starting material may not necessarily have been prepared from the description referred to. The Examples' stereochemistry has been assigned on the assumption that the absolute configuration centers are retained.
Reagents used in the following examples were commercially available from various suppliers (for example Sigma-Aldrich, Acros or Apollo scientific) and used without further purifications. Solvents were used in dry form. Reactions in anhydrous environment were run under a positive pressure of dry N2.
Microwave reactions were run on a Biotage Initiator 2.5 instrument.
Proton Nuclear Magnetic Resonance (1H NMR) spectra were recorded on Bruker Avance 400 MHz instrument. Chemical shifts are reported in ppm (δ) using the residual solvent line as internal standard. Splitting patterns are designated as: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; b, broad signal.
Mass spectra (MS) were run on a Ion Trap Thermo LCQ classic spectrometer, operating in positive ES(+) and negative ES(−) ionization mode.
UPLC spectra were performed on a Waters Acquity UPLC-SQD instrument using an Acquity UPLC-BEH C18 column (1.7 μM, 50×2.1 mm).
Flash silica gel chromatography was performed on Biotage automatic flash chromatography systems (Sp1 and Isolera systems) using Biotage SNAP HP silica cartridges or Biotage SNAP KP-NH cartridges.
Purifications of some basic compounds were performed using Phenomenex Strata SCX cartridges (55 μm, 70 A).
Thin layer chromatography was carried out using Merck TLC plates Kieselgel 60F-254, visualized with UV light, aqueous permanganate solution, iodine vapours.
The following abbreviations are used herein: DEAD:diethylazodicarboxylate; DIPEA: N,N-diisopropylethylamine; Boc: terbutyloxycarbonyl; DCM: dichloromethane; TFA: trifluoroacetic acid; DMF: dimethylformamide; THF: tetrahydrofuran; RT: room temperature; DMAP: dimethylamino pyridine; AcOEt: ethyl acetate.
Description 1: (S)-ethyl-6-oxopiperidine-2-carboxylate (D1)
Figure US09174977-20151103-C00013
Absolute ethanol (300 mL) was cooled at −5° C. then thionyl chloride (14.01 mL, 192.1 mmol) was added keeping the temperature below 0° C., followed by portionwise addiction of (S)-6-oxopiperidine-2-carboxylic acid (25.0 g, 174.6 mmol). The mixture was stirred at room temperature for 6 hours. The solvent was evaporated, then toluene (300 mL) and Et3N (48.7 mL) were added. After 0.5 hour the precipitate was filtered and washed with toluene and Et2O. The filtrate was evaporated and the residue treated with Et2O. The precipitate was eliminated and the solution was concentrated to give the title compound as yellow oil. Yield (29.9 g, 100%).
1H NMR (400 MHz, CDCl3): δ 1.30 (t, 3H), 1.75-1.95 (m, 3H), 2.15-2.25 (m, 1H), 2.30-2.45 (m, 2H), 4.08 (m, 1H), 4.24 (q, 2H), 6.43 (br s, 1H).
Description 2: (S)-1-tert-butyl-2-ethyl-6-oxopiperidine-1,2-dicarboxylate (D2)
Figure US09174977-20151103-C00014
To a solution of D1 (29.9 g, 174.6 mmol) in toluene (150 mL), DMAP (1.07 g, 8.73 mmol) was added followed by solution of Boc2O (45.74 g, 209.6 mmol) in toluene (100 mL). After 3.5 hours additional DMAP (20.0 g, 163.7 mmol) was added and the mixture was stirred at room temperature overnight. An aqueous solution of NaHCO3 (200 mL) was added and the separated organic layer was washed with water, dried over Na2SO4, filtered and concentrated. The residue was treated with cyclohexane (200 mL) and cooled with an ice bath, the precipitate was eliminated and the solution was concentrated to give a crude mixture which was purified by silica gel column chromatography (cyclohexane/ethyl acetate=6:4) to give the title compound as yellow oil. Yield (41.9 g, 88%).
1H NMR (400 MHz, CDCl3): δ 1.31 (t, 3H), 1.52 (s, 9H), 1.71-1.85 (m, 2H), 2.02-2.11 (m, 1H), 2.16-2.23 (m, 1H), 2.46-2.64 (m, 2H), 4.08 (m, 1H), 4.25 (m, 2H), 4.71 (dd, 1H). ESI+ m/z 565 [2M+Na]+ 294 [M+Na]+
Description 3: (S)-1-tert-butyl-2-ethyl-3,4-dihydropyridine-1,2(2H)-dicarboxylate (D3)
Figure US09174977-20151103-C00015
To a solution of D2 (20.98 g, 77.32 mmol) in toluene (200 mL) cooled at −50° C., LiEt3BH 1M in THF (81.2 mL, 81.19 mmol) was added keeping the temperature below −45° C. After 30 minutes at −45° C., DIPEA (57.9 mL, 332.5 mmol) was added followed by DMAP (0.142 g, 1.116 mmol) and trifluoroacetic anhydride (16 mL, 116 mmol). The mixture was stirred at room temperature for 2.5 hours and then cooled at 0° C. Water was added and the separated organic layer was washed with water, dried over Na2SO4, filtered and concentrated. The residue was dissolved in DCM (100 mL) and washed with 0.1 M aqueous solution of citric acid (150 mL). The solution was concentrated to give a crude mixture which was purified by silica gel column chromatography (cyclohexane/ethyl acetate=95:5) to give the title compound as orange oil. Yield (15.12 g, 77%).
1H NMR (400 MHz, CDCl3): δ 1.27 (m, 3H), 1.45-1.52 (m, 9H), 1.87-2.00 (m, 3H), 2.30-2.39 (m, 1H), 4.15-4.26 (m, 2H), 4.74-4.95 (m, 2H), 6.80-6.90 (m, 1H).
ESI+ m/z 156 [M+H−Boc]+
Description 4: (3S)-2-tert-butyl-3-ethyl-2-azabicyclo[4.1.0]heptane-2,3-dicarboxylate (D4)
Figure US09174977-20151103-C00016
A solution of D3 (20 g, 78.4 mmol) in toluene (400 mL) was cooled at −30° C., then Et2Zn 1M in hexanes (235 mL, 235 mmol) and a solution of CH2I2 (38 mL, 470 mmol) in toluene (50 mL) were added keeping the temperature below −30° C. The mixture was stirred at −15° C. for 16 hours; the temperature was raised to −5° C. then an aqueous solution of NaHCO3 was added. The separated organic layer was washed with brine, dried over Na2SO4, filtered and concentrated to give the crude mixture of diastereoisomers (28 g).
1H NMR (400 MHz, CDCl3): δ 0.22-0.54 (m, 1H), 0.75-0.92 (m, 1H), 1.14-1.32 (m, 4H), 1.45-1.51 (m, 9H), 1.65-2.07 (m, 4H), 2.60-3.03 (m, 1H), 4.12-4.58 (m, 3H).
ESI+ m/z 292 [M+Na]+
Description 5: (1R,3S,6S)-2-(tert-butoxycarbonyl)-2-azabicyclo[4.1.0]heptane-3-carboxylic acid (D5)
Figure US09174977-20151103-C00017
To a solution of D4 (25 g, 93 mmol) in ethanol (300 mL) cooled at 0° C., NaOH 2M (93 mL, 186 mmol) was added keeping the temperature below 5° C. The mixture was stirred at 0° C. for 2 hours and at room temperature for 3 hours and half. The solution was concentrated, water and DCM were added and the phases were separated.
The aqueous phase was washed with AcOEt, then i-Pr2O (the organic phases were collected, washed with water and concentrated to give a residue oil containing the diastereoisomeric mixture of unreacted esters that was used without any other purification in description 6); then the aqueous phase was acidified with acetic acid (pH=4), extracted with AcOEt, dried over Na2SO4, filtered and concentrated to give the title compound (D5) as a single diasteroisomer (white solid). Yield (11.0 g, 49%).
1H NMR (400 MHz, CDCl3): δ 4.63-4.43 (m, 1H), 2.99-2.90 (m, 1H), 2.10-1.87 (m, 2H), 1.77-1.61 (m, 2H), 1.52 (s, 9H), 1.29-1.19 (m, 1H), 0.94-0.81 (m, 1H), 0.33-0.25 (m, 1H).
Description 6: (3S)-2-(tert-butoxycarbonyl)-2-azabicyclo[4.1.0]heptane-3-carboxylic acid (D6)
Figure US09174977-20151103-C00018
The oil containing the diastereoisomeric mixture of unreacted esters from description 5 was dissolved in ethanol (300 mL) and NaOH 2M (90 mL, 180 mmol) was added. The mixture was stirred at 50° C. for 24 hours, then the solution was concentrated, water and i-Pr2O were added and the phases were separated. The aqueous phase was acidified with acetic acid (pH=4), extracted with DCM, dried over Na2SO4, filtered and concentrated to obtain the title compound (D6) as diasteroisomeric mixture (1/1 ratio) as yellow oil. Yield (8.0 g, 36%).
1H NMR (400 MHz, CDCl3): δ 4.63-4.25 (m, 1H), 2.99-2.71 (m, 1H), 2.13-1.62 (m, 4H), 1.52-1.46 (m, 9H), 1.28-1.18 (m, 1H), 0.92-0.76 (m, 1H), 0.51-0.25 (m, 1H).
Description 7: (1R,3S,6S)-tert-butyl-3-(hydroxymethyl)-2-azabicyclo[4.1.0]heptane-2-carboxylate (D7)
Figure US09174977-20151103-C00019
To a solution of D5 (11 g, 45.6 mmol) in THF (350 mL) cooled at 0° C., BH3 1M in THF (90 mL, 90 mmol) was added and the mixture was stirred at room temperature for 2 hours. Methanol (50 mL) was added; this solution was concentrated and co-evaporated twice from methanol to give the title compound. Yield (11 g, 100%).
1H NMR (400 MHz, CDCl3): δ 0.24 (m, 1H), 0.80-0.92 (m, 1H), 1.21 (m, 4H), 1.51 (s, 9H), 1.55-1.71 (m, 4H), 1.86 (m, 1H), 2.48-2.68 (m, 1H) 3.61-4.08 (m, 3H).
ESI+ m/z 250 [M+Na]+
Description 8: (1R,3S,6S)-tert-butyl-3-((1,3-dioxoisoindolin-2-yl)methyl)-2-azabicyclo[4.1.0]-heptane-2-carboxylate (D8)
Figure US09174977-20151103-C00020
A suspension of D7 (10 g, 44 mmol), phtalimide (10.29 g, 70 mmol) and triphenylphosphine (18.34 g, 70 mmol) in THF (150 mL) was cooled at 0° C., then a 40% solution of DEAD in toluene (31.94 mL, 70 mmol) was added. The mixture was stirred at room temperature for 3 hours, then water was added and the mixture was concentrated under vacuum; the residue was dissolved in DCM, washed with water and organics were evaporated. Cyclohexane (300 mL) and DCM (10 mL) were added, the precipitate was discarded and the filtrate was concentrated. The crude mixture was purified by silica gel column chromatography (cyclohexane/ethyl acetate=95:5 to 70/30) to give the title compound as a white solid. Yield (11 g, 71%).
1H NMR (400 MHz, CDCl3): δ 0.18-0.28 (m, 1H), 0.76-0.96 (m, 1H), 1.11-1.18 (m, 9H), 1.30-1.40 (m, 1H), 1.48-1.57 (m, 2H), 1.69-1.83 (m, 2H), 1.95-2.09 (m, 1H), 2.73-2.83 (m, 1H), 3.49-3.54 (m, 1H), 4.01-4.09 (m, 1H), 4.23-4.03 (m, 1H), 7.68 (m, 1H), 7.74 (m, 1H) 7.83-7.89 (m, 2H).
ESI+ m/z 735 [2M+Na]+
Description 9: (1R,3S,6S)-tert-butyl-3-(aminomethyl)-2-azabicyclo[4.1.0]heptane-2-carboxylate (D9)
Figure US09174977-20151103-C00021
To a solution of D8 (11 g, 30.9 mmol) in ethanol (200 mL) hydrazine hydrate (7.28 mL, 150 mmol) was added and the mixture was stirred at room temperature for 16 hours. The precipitate was filtered off and the filtrate was concentrated. Then i-Pr2O was added, the precipitate was discarded and the filtrate was concentrated to give the title compound as yellow oil. Yield (6.7 g, 96%).
1H NMR (400 MHz, CDCl3): δ 0.20-0.27 (m, 1H), 0.77-0.89 (m, 1H), 1.16 (m, 1H), 1.50 (m, 12H), 1.62-1.68 (m, 2H), 1.82-1.91 (m, 1H), 2.65-2.75 (m, 2H), 2.81-2.86 (m, 1H), 3.72-3.90 (m, 1H). ESI+ m/z 227 [M+Na]+
Description 10-13: (D10-13)
Figure US09174977-20151103-C00022

General Procedure 1
To a solution of D9 (4.5 mmol) in DMF (5 mL), K2CO3 (13.5 mmol) and Ar1-X (5.4 mmol) were added. The reaction mixture was heated at 120° C. until complete conversion of the starting material. The resulting mixture was poured in water and extracted with DCM. The organic layer was concentrated to obtain a crude mixture which was purified by silica gel chromatography (cyclohexane/ethyl acetate from 10/0 to 7/3) to obtain intermediate D10-D13 as detailed in below table.
According to general procedure 1 the following intermediates were prepared:
Intermediate Ar1 X MS Yield %
D10
Figure US09174977-20151103-C00023
F ESI+ m/z 372 [M + H]+ 87
D11
Figure US09174977-20151103-C00024
F ESI+ m/z 372 [M + H]+ 84
D12
Figure US09174977-20151103-C00025
F ESI+ m/z: 338 [M + H]+ 79
D13
Figure US09174977-20151103-C00026
Cl ESI+ m/z 373 [M + H] 88
Description 14-17 (D14-D17)
Figure US09174977-20151103-C00027

General Procedure 2
Intermediate D10-13 (1 eq.) were dissolved in dichloromethane (5 mL/mmol) and trifluoroacetic acid (2 mL/mmol) was added. After 1-16 hours at room temperature the volatiles were evaporated, the residue dissolved in dichloromethane and washed with saturated NaHCO3 aqueous solution. The organic layers were dried over Na2SO4, filtered and concentrated under vacuum to obtain intermediates D14-D17 as detailed in below table.
According to general procedure 2 the following intermediates were prepared:
Intermediate Ar1 MS 1H NMR Yield %
D14
Figure US09174977-20151103-C00028
ESI+ m/z 272 [M + H]+ 1HNMR (CDCl3) δ ppm 8.34 (bs, 1H), 7.57 (dd, J = 2, 8 Hz, 1H), 6.45 (d, J = 8 Hz, 1H), 5.44 (m, 1H), 3.33 (m, 2H), 2.86- 2.79 (m, 1H), 2.37 (m, 1H), 2.13 (m, 1H), 1.55-1.46 (m, 1H), 1.43-1.25 (m, 2H), 0.96 (m, 1H), 0.72-0.63 (m, 1H), 0.31-0.27 (m, 1H). 89
D15
Figure US09174977-20151103-C00029
ESI+ m/z 272 [M + H]+ 1HNMR (CDCl3) δ ppm 8.29 (bs, 1H), 7.53 (dd, J = 2, 8 Hz, 1H), 6.47 (d, J = 8 Hz, 1H), 6.13 (m, 1H), 3.62-3.47 (m, 2H), 3.10 (m, 1H), 2.59 (m, 1H), 2.22-2.11 (m, 1H), 1.78-1.68 (m, 1H), 1.66-1.60 (m, 1H), 1.57-1.47 (m, 1H), 1.18-1.09 (m, 1H), 0.86-0.80 (m, 1H), 0.61-0.57 (m, 1H). 100
D16
Figure US09174977-20151103-C00030
ESI+ m/z 238 [M + H]+ 1HNMR (CDCl3) δ ppm 10.17- 9.78 (bs, 1H), 7.90 (bs, 1H), 7.58 (dd, J = 2, 8 Hz, 1H), 6.86 (d, J = 8 Hz, 1H), 3.87 (m, 1H), 3.70 (m, 1H), 3.30-3.20 (m, 1H), 2.81 (m, 1H), 2.31-2.23 (m, 1H), 1.77-1.61 (m, 3H), 1.36-1.27 (m, 1H), 1.06-0.99 (m, 1H), 0.80-0.76 (m, 1H). 63
D17
Figure US09174977-20151103-C00031
ESI+ m/z 273 [M + H]+ 1HNMR (CDCl3) δ ppm 10.34 (bs, 1H), 9.44 (bs, 1H), 8.47 (bs, 1H), 7.81 (bs, 1H), 6.86 (d, J = 8 Hz, 1H), 3.88-3.84 (m, 1H), 3.50 (m, 1H), 2.77 (m, 1H), 2.30 (m, 1H), 1.73-1.51 (m, 4H), 1.25 (m, 1H), 0.95 (m, 1H), 0.68 (m, 1H). 100
Description 18: 2-bromo-5-cyanobenzoic acid (D18)
Figure US09174977-20151103-C00032
A solution of NaNO2 (0.29 g, 4.2 mmol) in water (1.6 mL) was added dropwise to a solution of 5-amino-2-bromobenzoic acid (0.86 g, 4 mmol) in HCl 2N (6 mL) and water (6 mL) at 0° C. during 15 minutes. The reaction mixture was stirred for 20 minutes and then added dropwise to a solution of CuCN (0.7 g, 8 mmol) and NaCN (0.4 g, 8 mmol) in water (5 mL) at 60° C.; the mixture was heated at 60° C. for further 15 minutes. After cooling at room temperature HCl (2N) was added and the product was extracted twice with AcOEt; the combined organic layers were dried and evaporated to give the title compound as a brown solid. Yield (0.6 g, 67%).
ESI− m/z 475 [2M+Na]
1H NMR (400 MHz, DMSO-d6): δ 7.89 (dd, 1H), 7.96 (d, 1H), 8.19 (d, 1H), 13.88 (br s, 1H).
Description 19-22 (D19-D22)
Figure US09174977-20151103-C00033

General Procedure 3
A suspension of D18 (91 mg, 0.4 mmol), N-methyl morpholine (150 μL; 1.36 mmol) and 2-chloro-4,6-dimethoxy-1,3,5-triazine (80 mg; 0.45 mmol) in dry 1,4-dioxane (1.5 mL) was stirred at 25° C. for 0.5 hours, then D14-D17 (0.4 mmol) dissolved in 1,4-dioxane (1.5 mL) were added. After 1-2 hours at 60-70° C., AcOEt and water were added; organics were dried over Na2SO4 and concentrated to a crude which was purified by silica gel column chromatography (DCM to DCM/MeOH 95/5) to obtain intermediate D19-D22 as detailed in below table.
According to general procedure 3 the following intermediates were prepared:
Intermediate Ar1 MS Yield %
D19
Figure US09174977-20151103-C00034
ESI+ m/z 480 [M + H]+ 73
D20
Figure US09174977-20151103-C00035
ESI+ m/z 480 [M + H]+ 63
D21
Figure US09174977-20151103-C00036
ESI+ m/z 446 [M + H]+ 73
D22
Figure US09174977-20151103-C00037
ESI+ m/z 481 [M + H]+ 65
Description 23: 5-chloro-2-iodobenzoyl chloride (D23)
Figure US09174977-20151103-C00038
To a solution of 5-chloro-2-iodobenzoic acid (3.0 g, 10.6 mmol) in toluene (150 mL), SOCl2 (7.75 mL, 106 mmol) was added and the mixture was heated at 100° C. for 3 hours. The solvent was concentrated in vacuum and the residue was co-evaporated from toluene twice to give the title compound as a grey solid. Yield (3.2 g, 100%)
1H NMR (400 MHz, CDCl3): δ 7.26 (dd, 1H), 7.98 (d, 1H), 8.03 (d, 1H).
Description 24: 5-methyl-2-iodobenzoyl chloride (D24)
Figure US09174977-20151103-C00039
To a solution of 5-methyl-2-iodobenzoic acid (3.0 g, 11.4 mmol) in toluene (150 mL), SOCl2 (8.35 mL, 114 mmol) was added and the mixture was heated at 100° C. for 3 hours. The solvent was concentrated in vacuum and the residue was co-evaporated from toluene twice to give the title compound as a grey solid. Yield (3.2 g, 100%)
1H NMR (400 MHz, CDCl3): δ 2.42 (s, 3H), 7.09 (dd, 1H), 7.90 (d, 1H), 7.92 (d, 1H).
Description 25-31: (D25-D31)
Figure US09174977-20151103-C00040

General Procedure 4
To a solution of intermediates D14-D17 (1 mmol) in DCM (2 mL) and triethylamine (2.2 mmol), a solution of D23-D24 (1 mmol) in DCM (2 mL) was added. The reaction mixture was stirred at room temperature until complete conversion of the starting material. The resulting mixture was washed with aqueous solution of NaHCO3, with water, dried and evaporated.
General Procedure 5
To a solution of intermediates D14-D17 (1 mmol) in DCM (6 mL) and triethylamine (2.2 mmol), a solution of D23-D24 (1.2 mmol) in DCM (2 mL) was added. The reaction mixture was stirred at room temperature until complete conversion of the starting material. The resulting mixture was washed with aqueous solution of NaHCO3, with water, dried and evaporated. Crude was purified by silica-NH chromatography (Cyclohexane/ethyl acetate from 10/0 to 5/5).
According to general procedure 4-5 the following intermediates were prepared:
Intermediate Ar1 R Procedure MS Yield %
D25
Figure US09174977-20151103-C00041
Cl 5 ESI+ m/z 536 [M + H]+ 19
D26
Figure US09174977-20151103-C00042
Me 4 ESI+ m/z 481 [M + H]+ 78
D27
Figure US09174977-20151103-C00043
Me 5 ESI+ m/z 517 [M + H]+ 29
D28
Figure US09174977-20151103-C00044
Cl 4 ESI+ m/z 536 [M + H]+ 76
D29
Figure US09174977-20151103-C00045
Cl 4 ESI+ m/z 503 [M + H]+ 66
D30
Figure US09174977-20151103-C00046
Cl 4 ESI+ m/z 537 [M + H]+ 72
Description 32-34: (D32-D34)
Figure US09174977-20151103-C00047

General Procedure 6
To a solution of D7 (1.32 mmol) in DMF (15 mL) cooled at 0° C., NaH 60% (1.58 mmol) was added. After stirring at room temperature for 10 minutes 2-F—Ar1 (1.58 mmol) was added and the reaction mixture was stirred at room temperature for 2-17 hours. An aqueous solution of NaHCO3 was added and the product was extracted with DCM, washed with brine, dried and concentrated to obtain a crude mixture which was purified by silica gel chromatography (cyclohexane/ethyl acetate from 10/0 to 7/3).
According to general procedure 6 the following intermediates were prepared:
Intermediate Ar1 MS Yield %
D32
Figure US09174977-20151103-C00048
ESI+ m/z 395 [M + Na]+ 73
D33
Figure US09174977-20151103-C00049
ESI+ m/z 361 [M + Na]+ 96
D34
Figure US09174977-20151103-C00050
ESI+ m/z 345 [M + Na]+ 47
Description 35-37: (D35-D37)
Figure US09174977-20151103-C00051

General Procedure 7
Intermediates D32-D34 (1 eq.) were dissolved in dichloromethane (10 ml/mmol) and trifluoroacetic acid (1.5 ml/mmol) was added. After 1 hour at room temperature the solution was evaporated, the residue dissolved in MeOH and loaded on a SCX cartridge, which was then washed with MeOH, followed by a solution of ammonia 2.0 M in MeOH. The basic fractions were collected and evaporated.
According to general procedure 7 the following intermediates were prepared:
Intermediate Ar1 MS 1HNMR Yield %
D35
Figure US09174977-20151103-C00052
ESI+ m/z 273 [M + H]+ 1HNMR (CDCl3) δ ppm 8.44 (bs, 1H), 7.78 (dd, J = 2, 8 Hz, 1H), 6.86 (d, J = 8 Hz, 1H), 4.30-4.28 (m, 2H), 3.07- 3.0 (m, 1H), 2.50-2.45 (m, 1H), 2.22- 2.14 (m, 1H), 1.68-1.61 (m, 2H, under water peak), 1.56-1.34 (m, 2H), 1.04- 0.96 (m, 1H), 0.65-0.59 (m, 1H), 0.36- 0.32 (m, 1H). 91
D36
Figure US09174977-20151103-C00053
ESI+ m/z 239 [M + H]+ 1HNMR (CDCl3) δ ppm 8.10 (bs, 1H), 7.53 (dd, J = 2, 8 Hz, 1H), 6.73 (d, J = 8 Hz, 1H), 4.22-4.16 (m, 2H), 3.03- 2.97 (m, 1H), 2.49-2.44 (m, 1H), 2.22- 2.13 (m, 1H), 1.91-1.78 (m, 1H,), 1.68-1.59 (m, 1H), 1.54-1.46(m, 1H), 1.41-1.32 (m, 1H), 1.04-0.95 (m, 1H), 0.63-0.57 (m, 1H), 0.36-0.32 (m, 1H). 86
D37
Figure US09174977-20151103-C00054
ESI+ m/z 223 [M + H] 1HNMR (CDCl3) δ ppm 7.98 (d, J = 2 Hz, 1H), 7.37-7.32 (m, 1H), 6.74 (m, 1H), 4.21-4.15 (m, 2H), 3.04-2.98 (m, 1H), 2.49-2.45 (m, 1H), 2.21-2.13 (m, 1H), 1.84 (m, 1H,), 1.69-1.60 (m, 1H), 1.54-1.47 (m, 1H), 1.42-1.32 (m, 1H), 1.03-0.95 (m, 1H), 0.63-0.57 (m, 1H), 0.36-0.33 (m, 1H). 79
Description 38-39 (D38-D39)
Figure US09174977-20151103-C00055
General procedure 8
A suspension of D18 (80 mg, 0.35 mmol), N-methyl morpholine (95 μl; 0.88 mmol) and 2-chloro-4,6-dimethoxy-1,3,5-triazine (56.5 mg; 0.32 mmol) in dry 1,4-dioxane (2 ml) was stirred at 25° C. for 1 hour, then (D36-D37) (0.29 mmol) dissolved in 1,4-dioxane (2 ml) were added. The reaction mixture was heated at 80° C. for 1.5 hours and concentrated in vacuum. The residue was dissolved in DCM, washed with an aqueous solution of NaHCO3, with an aqueous solution of NH4Cl, dried and concentrated to obtain a crude mixture which was purified by silica gel chromatography (cyclohexane/ethyl acetate 8/2).
According to general procedure 8 the following intermediates were prepared:
Intermediate Ar1 Procedure MS Yield %
D38
Figure US09174977-20151103-C00056
8 ESI+ m/z 447 [M + H]+ 63
D39
Figure US09174977-20151103-C00057
8 ESI+ m/z 431 [M + H]+ 54
Description 40-44: (D40-D44)
Figure US09174977-20151103-C00058

General Procedure 9
To a solution of intermediates D35-D37 (0.18 mmol) in DCM (1 mL) and thriethylamine (0.4 mmol), a solution of D23-D24 (0.18 mmol) in DCM (1 mL) was added. The reaction mixture was stirred at room temperature for 1-1.5 hours. The resulting mixture was washed with water, with aqueous solution of NaHCO3, dried and evaporated.
General Procedure 10
To a solution of intermediates D35-D37 (0.18 mmol) in DCM (1 mL) and thriethylamine (0.4 mmol), a solution of D23-D24 (0.18 mmol) in DCM (1 mL) was added. The reaction mixture was stirred at room temperature for 1-1.5 hours. The resulting mixture was washed with water, with aqueous solution of NaHCO3, dried and evaporated. The crude was purified by silica gel chromatography (cyclohexane/ethyl acetate 8/2).
According to general procedures 9-10 the following intermediates were prepared:
Intermediate Ar1 R Procedure MS Yield %
D40
Figure US09174977-20151103-C00059
Cl 9 ESI+ m/z 487 [M + H]+ 96
D41
Figure US09174977-20151103-C00060
Cl 9 ESI+ m/z 537 [M + H]+ 95
D42
Figure US09174977-20151103-C00061
Cl 9 ESI+ m/z 504 [M + H]+ 94
D43
Figure US09174977-20151103-C00062
Me 9 ESI+ m/z 483 [M + H]+ 75
D44
Figure US09174977-20151103-C00063
Me 10 ESI+ m/z 517 [M + H]+ 66
Description 45: (3S)-tert-butyl 3-(hydroxymethyl)-2-azabicyclo[4.1.0]heptane-2-carboxylate (D45)
Figure US09174977-20151103-C00064
To a solution of D6 (8 g, 45.6 mmol) in THF (250 mL) cooled at 0° C., BH3 1M in THF (66 mL, 66 mmol) was added and the mixture was stirred at room temperature for 2 hours. Methanol was added, the solution was concentrated in vacuum and co-evaporated twice from methanol to give the title compound as 1/1 diasteroisomeric mixture. Yield (7.6 g, 100%)
1H NMR (400 MHz, CDCl3): δ 3.59-4.07 (m, 3H), 2.46-2.89 (m, 1H), 1.82-2.07 (m, 1H), 1.54-1.70 (m, 4H), 1.51 (s, 9H), 0.94-1.22 (m, 1H), 0.18-0.82 (m, 1H).
ESI+ m/z 250 [M+Na]+
Description 46: (3S)-tert-butyl-3-((1,3-dioxoisoindolin-2-yl)methyl)-2-azabicyclo[4.1.0]-heptane-2-carboxylate (D46)
Figure US09174977-20151103-C00065
A suspension of D45 (7.6 g, 33.5 mmol), phtalimide (7.8 g, 53 mmol) and triphenylphosphine (13.9 g, 53 mmol) in THF (110 mL) was cooled at 0° C. then a 40% solution of DEAD in toluene (24 mL, 53 mmol) was added. The mixture was stirred at room temperature for 3 hours, then water was added and the mixture was concentrated under vacuum; the residue was dissolved in DCM, washed with water then organics were evaporated. Cyclohexane (237.5 mL) and DCM (12.5 mL) were added, the precipitate was discarded and the filtrate was concentrated to a crude mixture (12 g) which was used without any further purification.
ESI+ m/z 735 [2M+Na]+
Description 47: (3S)-tert-butyl 3-(aminomethyl)-2-azabicyclo[4.1.0]heptane-2-carboxylate (D47)
Figure US09174977-20151103-C00066
To a solution of D46 (12 g, 33 mmol) in ethanol (200 mL), hydrazine hydrate (7.3 mL, 150 mmol) was added and the mixture was stirred at room temperature for 16 hours. The precipitate was filtered off and the filtrate was concentrated. Then i-Pr2O was added, the precipitate was discarded and the filtrate was concentrated.
The residue was dissolved in MeOH and loaded on a SCX cartridge, which was then washed with MeOH, followed by an ammonia solution (2.0M in MeOH). The basic fractions were collected and evaporated to give a yellow oil as the title compound (disteroisomeric mixture). Yield (4.3 g, 58%)
1H NMR (400 MHz, CDCl3): δ 0.19-0.30 (m, 1H), 0.77-0.95 (m, 1H), 1.15-1.25 (m, 1H), 1.43-1.56 (m, 12H), 1.61-1.66 (m, 2H), 1.81-2.04 (m, 1H), 2.63-2.87 (m, 2H), 2.81-2.86 (m, 1H), 3.73-3.95 (m, 1H).
ESI+ m/z 227 [M+Na]+
Description 48-51: (D48-D51)
Figure US09174977-20151103-C00067

General Procedure 11
To a solution of D47 (4.86 mmol) in DMF (8 mL), K2CO3 (8.68 mmol) and Ar1-X (where X is 2-chloro or fluoro; 5.8 mmol) were added. The reaction mixture was heated at 80-130° C. until complete conversion of the starting material. The resulting mixture was poured into aqueous solution of NH4Cl and extracted with AcOEt. The organic layer was dried and concentrated to obtain a crude mixture which was purified by silica gel chromatography (cyclohexane/ethyl acetate from 10/0 to 8/2) to give the title compound as single diasteroisomer.
General Procedure 12
To a solution of D47 (6 mmol) in DMF (12 mL), K2CO3 (18 mmol) and Ar1-X (where X is 2-chloro or fluoro; 7.2 mmol) were added. The reaction mixture was heated at 120° C. until complete conversion of the starting material. The resulting mixture was poured in water and extracted with DCM. The organic layer was concentrated to obtain a crude mixture which was purified by silica gel chromatography (cyclohexane/ethyl acetate from 10/0 to 75/25) to give the title compound as pure diasteroisomer.
According to general procedures 11-12 the following intermediates were prepared:
Intermediate Ar1 X Procedure MS Yield %
D48
Figure US09174977-20151103-C00068
F 12 ESI+ m/z 372 [M + H]+ 38
D49
Figure US09174977-20151103-C00069
F 11 ESI+ m/z 372 [M + H]+ 31
D50
Figure US09174977-20151103-C00070
F 11 ESI+ m/z 338 [M + H]+ 17
D51
Figure US09174977-20151103-C00071
Cl 11 ESI+ m/z 373 [M + H]+ 25
Description 52-55
Figure US09174977-20151103-C00072

General Procedure 13
Intermediates D48-D51 (1 eq.) were dissolved in dichloromethane (3 mL/mmol) and trifluoroacetic acid (1 mL/mmol) was added. After 1.5 hours at room temperature the solution was diluted with MeOH and loaded on a SCX cartridge, which was then washed with MeOH, followed by an ammonia solution (2.0M in MeOH). The basic fractions were collected and evaporated.
General Procedure 14
Intermediates D48-D51 (1 eq.) were dissolved in dichloromethane (4 mL/mmol) and trifluoroacetic acid (2 mL/mmol) was added. After 2 hours at room temperature the solution was evaporated, the residue dissolved in dichloromethane and washed with saturated NaHCO3 aqueous solution. The organic layers were dried (Na2SO4) and concentrated under vacuum.
According to general procedures 13-14 the following intermediates were prepared:
Intermediate Ar1 Procedure MS 1HNMR Yield %
D52
Figure US09174977-20151103-C00073
14 ESI+ m/z 273 [M + H]+ 1HNMR (CDCl3) δ ppm 8.33 (bs, 1H), 7.56 (dd, J = 2, 8 Hz, 1H), 6.44 (d, J = 8 Hz, 1H), 5.32 (m, 1H), 3.46-3.40 (m, 1H), 3.05-2.98 (m, 1H), 2.77-2.70 (m, 1H), 2.51-2.46 (m, 1H), 2.09-1.91 (m, 2H), 1.43-1.21 (m, 1H), 1.06-0.94 (m, 2H), 0.78-0.72 (m, 1H), 0.27-0.23 (m, 1H). 74
D53
Figure US09174977-20151103-C00074
13 ESI+ m/z 273 [M + H]+ 1HNMR (CDCl3) δ ppm 8.21 (d, J = 2 Hz, 1H), 6.73 (d, J = 8Hz, 1H), 6.59 (s, 1H), 5.19 (m, 1H), 3.45-3.38 (m, 1H), 3.03-2.96 (m, 1H), 2.76- 2.70 (m, 1H), 2.50-2.46 (m, 1H,), 2.08-1.92 (m, 2H), 1.61- 1.56 (m, 1H), 1.06-0.94 (m, 2H), 0.77-0.72 (m, 1H), 0.26- 0.22 (m, 1H). 70
D54
Figure US09174977-20151103-C00075
13 ESI+ m/z 238 [M + H]+ 1HNMR (CDCl3) δ ppm 8.02 (d, J = 2 Hz, 1H), 7.34 (dd, J = 2, 8 Hz, 1H), 8.37 (d, J = 8 Hz, 1H), 4.91 (m, 1H), 3.36-3.30 (m, 1H), 2.98-2.92 (m, 1H), 2.74- 2.67 (m, 1H), 2.50-2.45 (m, 1H,), 2.07-1.90 (m, 2H), 1.59- 1.54 (m, 1H, under water peak), 1.04-0.93 (m, 2H), 0.76-0.70 (m, 1H), 0.25-0.21 (m, 1H). 100
D55
Figure US09174977-20151103-C00076
13 ESI+ m/z 274 [M + H]+ 1HNMR (CDCl3) δ ppm 8.48 (bs, 1H), 6.81 (d, J = 2 Hz, 1H), 5.79 (m, 1H), 3.53-3.47 (m, 1H), 3.17-3.10 (m, 1H), 2.76- 2.69 (m, 1H), 2.50-2.46 (m, 1H), 2.07-1.90 (m, 2H), 1.60- 1.54 (m, 1H, under water peak), 1.04-0.91 (m, 1H), 0.75-0.70 (m, 1H), 0.25-0.22 (m, 1H). 77
Description 56: 4-bromo-3-((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile (D56)
Figure US09174977-20151103-C00077
A suspension of D18 (68 mg, 0.3 mmol;), N-methyl morpholine (110 μL; 1.02 mmol) and 2-chloro-4,6-dimethoxy-1,3,5-triazine (60 mg; 0.34 mmol) in dry 1,4-dioxane (1 mL) was stirred at 25° C. for 0.5 hour, then D52 (0.3 mmol) dissolved in 1,4-dioxane (1 mL) was added. After 1 hour at 60° C., DCM and water were added. The organic layer was separated, washed with aqueous citric acid solution, washed with saturated NaHCO3 aqueous solution and concentrated in vacuum.
ESI+ m/z 480 [M+H]+
Description 57-64: (D57-D64)
Figure US09174977-20151103-C00078

General Procedure 15
To a solution of intermediates D52-D55 (0.085 mmol) in DCM (1 mL) and triethylamine (0.17 mmol), a solution of D23-D24 (0.1 mmol) in DCM (1 mL) was added. The reaction mixture was stirred at room temperature for 2 hours then washed with water, dried and evaporated. Crude was purified by silica gel column chromatography (Cyclohexane/ethyl acetate from 10/0 to 5/5).
General Procedure 16
To a solution of intermediates D52-D55 (0.3 mmol) in DCM (5 mL) and triethylamine (0.45 mmol), a solution of D23-D24 (0.3 mmol) in DCM (2 mL) was added. The reaction mixture was stirred at room temperature for 0.5 hours then washed with water, dried and evaporated. Crude was purified by silica gel column chromatography (Cyclohexane/ethyl acetate from 10/0 to 5/5).
General Procedure 17
To a suspension of intermediates D52-D55 (0.25 mmol) and Si-diethylamine (silica supported reagent, Silicycle, loading 1.25 mmol/g, 300 mg, 0.375 mmol) in DCM (0.5 mL), a solution of D23-D24 (0.25 mmol) in DCM (0.5 mL) was added. The reaction mixture was stirred at room temperature for 2-48 hours then filtered, washed with methanol/DCM 1/1. The solution was loaded on a SCX cartridge, which was then washed with MeOH, followed by an ammonia solution (2.0M in MeOH). The ammonia eluted fractions were collected and evaporated to give the title compounds.
According to general procedure 15-17 the following intermediates were prepared:
Intermediate Ar1 R Procedure MS Yield %
D57
Figure US09174977-20151103-C00079
Me 15 ESI+ m/z 516 [M + H]+ 98
D58
Figure US09174977-20151103-C00080
Cl 16 ESI+ m/z 535 [M + H]+ 87
D59
Figure US09174977-20151103-C00081
Me 17 ESI+ m/z 515 [M + H]+ 90
D60
Figure US09174977-20151103-C00082
Cl 17 ESI+ m/z 502 [M + H]+ 84
D61
Figure US09174977-20151103-C00083
Cl 17 ESI+ m/z 535 [M + H]+ 88
D62
Figure US09174977-20151103-C00084
Me 17 ESI+ m/z 481 [M + H]+ 81
D63
Figure US09174977-20151103-C00085
Cl 17 ESI+ m/z 536 [M + H]+ 46
D64
Figure US09174977-20151103-C00086
Me 17 ESI+ m/z 516 [M + H]+ 54
Description 65: 2-methyl-5-phenylthiazole-4-carboxylic acid (D65)
Figure US09174977-20151103-C00087
2-methyl-5-phenylthiazole-4-carboxylic acid may be prepared as in the procedure described in U.S. Pat. No. 3,282,927.
EXAMPLES Example 1 Preparation of Compounds 1a-k
Figure US09174977-20151103-C00088

General Procedure 18
Intermediates (D19-22 and D25-31) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was warmed at 130° C. for 10 minutes (microwave), then poured in aqueous saturated solution of NH4Cl and extracted with AcOEt (3×50 ml). The organic layers were combined, dried (Na2SO4) and concentrated under vacuum; the crude mixture was purified by silica gel column chromatography (Cyclohexane 100% to Cyclohexane/Acetone 8/2 or Cyclohexane 100% to cyclohexane/AcOEt 2/8) to give the title compounds.
General Procedure 19
Intermediates (D19-22 and D25-31) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was warmed at 130° C. for 10 minutes (microwave), then at 90° C. for 18 hours, then for further 8 hours at 120° C. The reaction mixture was poured in aqueous saturated solution of NH4Cl and extracted with AcOEt; the organic layers were combined, dried (Na2SO4) and concentrated under vacuum. The residue was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by a solution of ammonia 2.0 M in MeOH. The basic fractions were collected and evaporated. The residue was purified by silica-NH column chromatography (Cyclohexane 100% to AcOEt 100%) to give the title compounds.
General Procedure 20
Intermediates (D19-22 and D25-31) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was warmed at 100° C. for 20 minutes (microwave), then poured in aqueous saturated solution of NH4Cl and extracted with DCM. The organic layers were combined, dried (Na2SO4) and concentrated under vacuum; the residue was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by a solution of ammonia 2.0 M in MeOH. The basic fractions were collected and evaporated. The residue was purified by silica gel column chromatography (Cyclohexane 100% to Cyclohexane/Acetone 7/3) to give the title compounds.
General Procedure 21
Intermediates (D19-22 and D25-31) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was warmed at 130° C. for 10 minutes (microwave). The reaction mixture was poured in water and extracted with DCM. The organic layers were combined, dried (Na2SO4) and concentrated under vacuum then purified by silica-NH column chromatography (Cyclohexane 100% to cyclohexane/AcOEt 2/8) to give a residue that was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by a solution of ammonia 2.0 M in MeOH. The basic fractions were collected and evaporated to give the title compounds.
General Procedure 22
Intermediates (D19-22 and D25-31) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was warmed at 130° C. for 10 minutes (microwave), then poured in water and extracted with DCM. The organic layers were combined, dried (Na2SO4) and concentrated under vacuum; the crude mixture was purified by silica gel column chromatography (Cyclohexane 100% to cyclohexane/Acetone 7/3) to give a residue that was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by an ammonia solution (2.0M in MeOH). The basic fractions were collected and evaporated to give the title compounds.
Compounds 1a-k were prepared according to general procedure 18-22:
Comp. Intermediate Procedure Yield %
1a (D25) 18 39
Figure US09174977-20151103-C00089
1H NMR (Acetone-d6) δ ppm = 8.89-8.80 (m, 2 H), 8.36-8.01 (m, 2 H), 7.57-7.35 (m, 3 H), 6.80-6.77 (m, 2 H), 4.62 (m, 1 H), 3.82- 3.72 (m, 2 H), 2.96-2.62 (m, 1 H), 2.22-2.10 (m, 1 H), 1.91-1.82 (m, 1 H), 1.77-1.64 (m, 1H), 1.44-1.25 (m, 2H), 0.61-0.38 (m, 2H). ESI+ m/z 488 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
1b (D19) 22   5
Figure US09174977-20151103-C00090
1H NMR (CDCl3) δ ppm 8.66-8.62 (m, 2H), 8.54-8.52 (m, 1H), 8.40 (m, 1H), 7.88-7.81 (m, 2H), 7.59-7.56 (m, 1H), 7.51-7.46 (m, 1H), 7.17-7.12 (m, 1H), 4.77-4.70 (m, 1H), 3.87-3.78 (m, 1H), 3.76-3.57 (m, 2H), 2.61-2.53 (m, 1H), 2.16-2.01 (m, 1H), 1.95- 1.78 (m, 2H), 1.75-1.62 (m, 1H), 1.36-1.28 (m, 1H), 0.63-0.17 (m, 2H). ESI+ m/z 479 [M + H]+
4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile
1c (D26) 18 11
Figure US09174977-20151103-C00091
1HNMR (CDCl3) δ ppm 8.86-8.62 (m, 2H), 8.29-8.27 (m, 1H), 8.07 (m, 1H), 7.33-7.31 (m, 2H), 7.07 (m, 2H), 6.46-6.34 (m, 1H), 5.93-5.75 (m, 1H), 4.76-4.74 (m, 1H), 3.76 (m, 1H), 3.61-3.42 (m, 1H), 2.49-2.35 (m, 5H), 2.07-1.99 (m, 1H), 1.74-1.22 (m, 2H), 1.33-1.09 (m, 1H), 0.58-0.05 (m, 2H) ESI+ m/z 434 [M + H]+
((1R,3S,6S-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-
(pyrimidin-2-yl)phenyl)methanone
1d (D27) 21 18
Figure US09174977-20151103-C00092
1H NMR (Acetone-d6) δ ppm = 8.85-8.77 (m, 2 H), 8.36-8.09 (m, 2 H), 7.67-7.54 (m, 1 H), 7.28-7.25 (m, 2 H), 6.67-6.47 (m, 2 H), 4.64 (m, 1 H), 3.82-3.63 (m, 2 H), 2.96-2.61 (m, 1 H), 3.43-2.31 (m, 3 H), 2.18-2.10 (m, 1H), 1.91-1.82 (m, 1H), 1.73-1.61 (m, 2H), 1.42-1.29 (m, 2H), 0.57- 0.27 (m, 2H). ESI+ m/z 468 [M + H]+
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
1e (D28) 21 11
Figure US09174977-20151103-C00093
1H NMR (Acetone-d6) δ ppm = 8.89-8.82 (m, 2 H), 8.37-8.17 (m, 2 H), 7.67-7.57 (m, 2 H), 7.46-7.37 (m, 2 H), 6.68-6.54 (m, 1 H), 4.62 (m, 1 H), 3.80-3.68 (m, 2H), 2.95-2.82 (m, 1H), 2.22-2.10 (m, 1H), 1.91-1.82 (m, 1H), 1.75-1.68 (m, 2H), 1.48-1.34 (m, 2H), 0.80-0.31 (m, 2H). ESI+ m/z 488 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
1f (D29) 21 29
Figure US09174977-20151103-C00094
1H NMR (Acetone-d6) δ ppm = 8.89-8.82 (m, 2 H), 8.36-8.19 (m, 1 H), 8.02-7.74 (m, 1 H), 7.59-7.31 (m, 3 H), 6.58-6.43 (m, 1 H), 6.24-6.14 (m, 1 H), 4.60 (m, 1H), 3.72-3.60 (m, 2H), 2.95-2.60 (m, 1H), 2.18-2.09 (m, 1H), 1.88-1.84 (m, 1H), 1.73-1.63 (m, 2H), 1.40-1.32 (m, 1H), 1.22-0.9 (m, 1H), 0.63-0.33 (m, 2H). ESI+ m/z 455 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
1g (D30) 21 14
Figure US09174977-20151103-C00095
1H NMR (Acetone-d6) δ ppm = 8.90-8.83 (m, 2 H), 8.69-8.58 (m, 1 H), 8.38-8.35 (m, 1 H), 7.59-7.37 (m, 2 H), 7.13-6.90 (m, 2 H), 4.70 (m, 1H), 3.84-3.71 (m, 2H), 2.98-2.65 (m, 1H), 2.20-2.10 (m, 1H), 1.91-1.84 (m, 1H), 1.78-1.70 (m, 2H), 1.40-1.29 (m, 2H), 0.60-0.36 (m, 2H). ESI+ m/z 489 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-
yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone
1h (D20) 21  6
Figure US09174977-20151103-C00096
1H NMR (Acetone-d6) δ ppm = 8.95-8.87 (m, 2 H), 8.51-8.35 (m, 1 H), 8.29-8.27 (m, 1 H), 7.97-7.84 (m, 2 H), 7.52-7.43 (m, 1 H), 6.80-6.77 (m, 1 H), 6.71-6.45 (m, 1H), 4.64 (m, 1H), 3.86-3.68 (m, 2H), 2.96-2.65 (m, 1H), 2.22-2.09 (m, 1H), 1.91-1.86 (m, 1H), 1.77-1.67 (m, 2H), 1.44-1.31 (m, 2H), 0.63-0.41 (m, 2H). ESI+ m/z 479 [M + H]+
4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile
1i (D21) 19  7
Figure US09174977-20151103-C00097
1HNMR (CDCl3) δ ppm 8.83-8.82 (m, 2H), 8.58-8.56 (m, 1H), 7.87-7.72 (m, 4H), 7.52-7.49 (m, 1H), 7.33-7.30 (m, 1H), 4.48- 4.38 (m, 1H), 3.83-3.80 (m, 1H), 3.60-3.54 (m, 1H), 2.49-2.45 (m, 1H), 2.19-2.13 (m, 1H), 1.93-1.88 (m, 1H), 1.75-1.64 (m, 2H), 1.33- 1.26 (m, 1H), 0.59-0.15 (m, 2H). ESI+ m/z 445 [M + H]+
3-((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)-4-
(pyrimidin-2-yl)benzonitrile
1j (D22) 19 10
Figure US09174977-20151103-C00098
1HNMR (CDCl3) δ ppm 8.85-8.84 (m, 2H), 8.56-8.34 (m, 2H), 7.82-7.73 (m, 1H), 7.54-7.71 (m, 1H), 6.89-6.83 (m, 1H), 6.22-6.12 (m, 1H), 4.77 (m, 1H), 3.72-3.89 (m, 2H), 2.57-2.47 (m, 1H), 2.18- 2.06 (m, 1H), 1.91-1.77 (m, 2H), 1.69-1.60 (m, 1H), 1.48-1.28 (m, 2H), 0.64-0.21 (m, 2H). ESI+ m/z 480 [M + H]+
4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile
1k (D31) 20 18
Figure US09174977-20151103-C00099
1H NMR (Acetone-d6) δ ppm = 8.85-8.78 (m, 2 H), 8.69-8.57 (m, 1 H), 8.27-8.10 (m, 1 H), 7.39-7.34 (m, 1 H), 7.31-7.27 (m, 1 H), 7.13-7.01 (m, 1 H), 6.99-6.89 (m, 1H), 4.72 (m, 1H), 3.82-3.74 (m, 2H), 2.64-2.57 (m, 1H), 2.38-2.43 (m, 3H), 2.16- 2.09 (m, 1H), 1.91-1.86 (m, 1H), 1.76-1.65 (m, 2H), 1.42-1.31 (m, 2H), 0.59-0.33 (m, 2H). ESI+ m/z 469 [M + H]+
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-
yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone
Example 2 Preparation of Compounds 2a-c
Figure US09174977-20151103-C00100

General Procedure 23
D65 (0.06 mmol), N-methyl morpholine (0.20 mmol) and 2-chloro-4,6-dimethoxy-1,3,5-triazine (0.06 mmol) dissolved in dry 1,4-dioxane (0.5 mL) were stirred at 25° C. for 0.5 hours, then (D14-D17) (0.06 mmol) dissolved in 1,4-dioxane (0.5 mL) were added. After 2-16 hours at 60° C. the crude reaction mixture was purified by silica gel column chromatography (Cyclohexane to DCM/MeOH=9/1).
General Procedure 24
D65 (0.1 mmol), N-methyl-morpholine (0.30 mmol) and 2-chloro-4,6-dimethoxy-1,3,5-triazine (0.1 mmol) dissolved in dry 1,4-dioxane (0.5 mL) were stirred at 25° C. for 0.5 hours, then (D14-D17) (0.06 mmol) dissolved in 1,4-dioxane (0.5 mL) were added. After 2 hours at 60° C. the reaction mixture was diluted with DCM, washed with water and concentrated. The crude was purified by silica gel column chromatography (Cyclohexane to AcOEt or DCM to DCM/MeOH 95/5).
Compounds 2a-c were prepared according to general procedures 23-24:
Comp. Intermediate Procedure Yield %
2a (D14) 23 78
Figure US09174977-20151103-C00101
1H NMR (CDCl3) δ ppm 8.25-8.36 (m, 1H), 7.22-7.58 (m, 6H), 6.42-6.54 (m, 1H), 5.85-5.96 (m, 1H), 4.73-4.78 (m, 1H), 3.64- 3.74 (m, 1H), 3.42-3.54 (m, 1H), 3.07-3.31 (m, 1H), 2.59-2.74 (m, 3H), 1.64-2.38 (m, 2H), 1.49-1.57 (m, 1H), 1.30-1.40 (m, 1H), 1.06-1.15 (m, 1H), 0.39-0.69 (m, 1H), −0.04-0.24 (m, 1H). ESI+ m/z 473 [M + H]+
(2-methyl-5-phenylthiazol-4-yl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
2b (D16) 24 36
Figure US09174977-20151103-C00102
1H NMR (CDCl3) δ ppm 7.91 -8.02 (m, 1H), 7.48 (m, 1H), 7.36- 7.45 (m, 3H), 7.24-7.33 (m, 2H), 6.30-6.58 (m, 1H), 5.58-5.80 (m, 1H), 4.03-4.70 (m, 1H), 3.55-3.70 (m, 1H), 3.30-3.42 (m, 1H), 3.07-3.22 (m, 1H), 2.61-2.74 (m, 3H), 1.80-2.33 (m, 1H), 1.59-1.69 (m, 1H), 1.40-1.55 (m, 1H), 1.32-1.36 (m, 1H), 1.04- 1.15 (m, 1H), 0.36-0.59 (m, 1H), −0.07-0.22 (m, 1H). ESI+ m/z 439 [M + H]+
((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(2-methyl-
5-phenylthiazol-4-yl)methanone
2c (D17) 24 53
Figure US09174977-20151103-C00103
1H NMR (CDCl3) δ ppm 8.44-8.50 (m, 1H), 7.28-7.52 (m, 5H), 6.80-6.84 (m, 1H), 4.77-6.12 (m, 1H), 3.47-3.74 (m, 2H), 3.10- 3.31 (m, 1H), 2.63-2.74 (m, 3H), 1.88-2.33 (m, 2H), 1.25-1.67 (m, 3H), 1.08 (m, 1H), 0.39-0.89 (m, 1H), −0.02-0.24 (m, 1H). ESI+ m/z 474 [M + H]+
(2-methyl-5-phenylthiazol-4-yl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyrimidin-2-
yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone
Example 3 Preparation of Compounds 3a-q
Figure US09174977-20151103-C00104

General Procedure 25
Intermediates (D38-D44) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was heated at 100° C. for 10-20 minutes (microwave), then poured in water and extracted with DCM; the organic layers were combined, dried (Na2SO4) and concentrated under vacuum. The crude mixture was purified by silica gel column chromatography (Cyclohexane 100% to cyclohexane/AcOEt=1/1 or cyclohexane 100% to cyclohexane/Acetone=8/2) to give a residue that was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by a solution of ammonia 2.0 M in MeOH. The ammonia eluted fractions were collected and evaporated to give the title compounds.
General Procedure 26
Intermediates (D38-D44) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was heated at 100° C. for 30 minutes (microwave), then at 120° C. for 18 hours. The reaction mixture was poured in water and extracted with DCM; the organic layers were combined, dried (Na2SO4) and concentrated under vacuum. The residue was purified by silica gel column chromatography (Cyclohexane 100% to Cyclohexane/AcOEt=1/1) to give the title compounds.
Compounds 3a-g were prepared according to general procedure 25-26:
Comp. Intermediate Procedure Yield %
3a (D38) 25 21
Figure US09174977-20151103-C00105
1H NMR (Acetone-d6) δ ppm = 9.02-8.96 (m, 2 H), 8.52-8.41 (m, 1 H), 8.28-8.26 (m, 1 H), 8.0-7.90 (m, 2 H), 7.78-7.75 (m, 1 H), 7.56-7.53 (m, 1 H), 6.92-6.88 (m, 1 H), 4.74-4.72 (m, 1 H), 4.65- 4.61 (m, 1 H), 4.52-4.47 (m, 1H), 2.70-2.61 (m, 1H), 2.03-1.95 (m, 1H), 1.86-1.69 (m, 2H), 1.45-1.31 (m, 2H), 0.68-0.43 (m, 2H). ESI+ m/z 446 [M + H]+
3-((1R,3S,6S)-3-(((5-chloropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)-4-
(pyrimidin-2-yl)benzonitrile
3b (D39) 26 16
Figure US09174977-20151103-C00106
1H NMR (Acetone-d6) δ ppm = 9.02-8.96 (m, 2 H), 8.52-8.41 (m, 1 H), 8.18-8.16 (m, 1 H), 7.98-7.90 (m, 2 H), 7.63-7.58 (m, 1 H), 7.55- 7.52 (m, 1 H), 6.90-6.87(m, 1 H), 4.77-4.70 (m, 1 H), 4.63-4.59 (m, 1 H), 4.50-4.45 (m, 1H), 2.67-2.60 (m, 1H), 2.03-1.95 (m, 1H), 1.80- 1.66 (m, 2H), 1.41-1.31 (m, 2H), 0.67-0.44 (m, 2H). ESI+ m/z 430 [M + H]+
3-((1R,3S,6S)-3-(((5-fluoropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)-4-(pyrimidin-
2-yl)benzonitrile
3c (D40) 25 25
Figure US09174977-20151103-C00107
1HNMR (Acetone-d6) δ ppm 8.96-8.90 (m, 2H), 8.37-8.26 (m, 1H), 8.17-7.91 (m, 1H), 7.63-7.56 (m, 2H), 7.47-7.45 (dd, 2H), 6.90- 6.87 (m, 1H), 4.74-4.70 (m, 1H), 4.65-4.60 (m, 1H), 4.48-4.25 (m, 1H), 2.95-2.58 (m, 1H), 2.04-1.97 (m, 1H), 1.81-1.67 (m, 2H), 0.61- 0.42 (m, 4H). ESI+ m/z 439 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-fluoropyridin-2-yl)oxy)methyl)-2-azabicyclo-
[4.1.0]heptan-2-yl)methanone
3d (D41) 25 20
Figure US09174977-20151103-C00108
1H NMR (CDCl3) δ ppm = 8.87-8.80 (m, 2 H), 8.52-8.34 (m, 1 H), 8.39-8.28 (m, 1 H), 7.83-7.80 (m, 1 H), 7.55-7.38 (m, 2 H), 7.24- 7.22 (m, 1 H), 6.90-6.87 (m, 1 H), 4.93-4.87 (m, 1 H), 4.71-4.67 (m, 1 H), 4.57-4.25 (m, 1H), 2.54-2.49 (m, 1H), 2.06-1.99 (m, 2H), 1.78- 1.70 (m, 1H), 1.31-1.12 (m, 2H), 0.62-0.27 (m, 2H). ESI+ m/z 489 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)oxy)methyl)-2-aza-
bicyclo[4.1.0]heptan-2-yl)methanone
3e (D42) 25 60
Figure US09174977-20151103-C00109
1H NMR (CDCl3) δ ppm = 8.86-8.79 (m, 2 H), 8.38-8.28 (m, 1 H), 8.18-7.99 (m, 1 H), 7.58-7.55 (m, 1 H), 7.50-7.47 (m, 1 H), 7.37 (m, 1 H), 7.23-7.21 (m, 1H), 6.80-6.75 (m, 1H), 4.91-4.84 (m, 1H), 4.62- 4.54 (m, 1H), 4.48-4.43 (m, 1H), 2.52-2.48 (m, 1H), 2.07-1.97 (m, 2H), 1.78-1.69 (m, 1H), 1.44-1.20 (m, 2H), 0.62-0.24 (m, 2H). ESI+ m/z 456 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-chloropyridin-2-yl)oxy)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
3f (D43) 25 37
Figure US09174977-20151103-C00110
1H NMR (CDCl3) δ ppm = 8.84-8.78 (m, 2 H), 8.30-8.21 (m, 1 H), 8.17-7.94 (m, 1 H), 7.58-7.51(m, 1 H), 7.35-7.31 (m, 1 H), 7.18- 7.15 (m, 2 H), 6.78-6.63 (m, 1H), 4.92 (m, 1H), 4.63-4.59 (m, 1H), 4.46-4.41 (m, 1H), 2.50-2.40 (m, 4H), 2.04-1.98 (m, 2H), 1.81-1.65 (m, 1H), 1.43-1.17 (m, 2H), 0.62-0.11 (m, 2H). ESI+ m/z 435 [M + H]+
((1R,3S,6S)-3-(((5-chloropyridin-2-yl)oxy)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-(pyrimidin-
2-yl)phenyl)methanone
3g (D44) 25 46
Figure US09174977-20151103-C00111
1H NMR (CDCl3) δ ppm = 8.85-8.78 (m, 2 H), 8.52 (m, 1 H), 8.31- 8.21 (m, 1 H), 7.83-7.76 (m, 1 H), 7.34-7.30 (m, 1 H), 7.20-7.17 (m, 2 H), 6.90-6.75 (m, 1H), 4.98-4.90 (m, 1H), 4.72-4.69 (m, 1H), 4.56- 4.51 (m, 1H), 2.51-2.39 (m, 4H), 2.06-1.96 (m, 2H), 1.82-1.67 (m, 1H), 1.43-1.18 (m, 2H), 0.63-0.10 (m, 2H). ESI+ m/z 435 [M + H]+
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((5-(trifluoromethyl)pyridin-2-yl)oxy)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
Example 4 Preparation of Compounds 4a-i
Figure US09174977-20151103-C00112

General Procedure 27
Intermediates (056-064) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was heated at 130° C. for 10 minutes (microwave), then poured in water and extracted with DCM; the organic layers were combined, dried (Na2SO4) and concentrated under vacuum. The crude mixture was purified by silica gel column chromatography (Cyclohexane 100% to Cyclohexane/Acetone 7/3) to give a residue that was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by an ammonia solution (2.0M in MeOH). The ammonia eluted fractions were collected and evaporated to give the title compounds.
General Procedure 28
Intermediates (056-064) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.5 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was heated at 130° C. for 10 minutes (microwave), then poured in water and extracted with DCM; the organic layers were combined, dried (Na2SO4) and concentrated under vacuum. The crude mixture was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by an ammonia solution (2.0M in MeOH). The basic fractions were collected and evaporated; the residue was purified by silica gel column chromatography (Cyclohexane 100% to cyclohexane/AcOEt=1/1) to give the title compounds.
General Procedure 29
Intermediates (056-064) (1 mmol) were dissolved in dry DMF (15 ml/mmol) under nitrogen atmosphere, then CsF (2 mmol), CuI (0.2 mmol), [Ph3P]4Pd (0.1 mmol) and pyrimidine-2-tributylstannane (1.2 mmol; prepared according to Eur. J. Org. Chem. 2003, 1711-1721) were added. The mixture was heated at 120° C. for 18 hours, then poured in aqueous saturated solution of NaHCO3 and extracted with DCM. The organic layers were filtered over a celite pad, dried (Na2SO4) and concentrated under vacuum; the crude mixture was dissolved in MeOH then loaded on a SCX cartridge, which was then washed with MeOH, followed by an ammonia solution (2.0M in MeOH). The basic fractions were collected and evaporated; the residue was purified by silica-NH column chromatography (Cyclohexane 100% to Cyclohexane/AcOEt=1/1) to give the title compounds.
Compounds 4a-i were prepared according to general procedure 27-29:
Comp. Intermediate Procedure Yield %
4a (D57) 28 32
Figure US09174977-20151103-C00113
1H NMR (CDCl3) δ ppm = 8.64 (br. s., 2 H), 8.35-8.26 (m, 2 H), 7.56-7.54 (m, 1 H),7.35-7.31 (m, 1H), 7.30-7.13 (m, 2 H, under the solvent peak), 6.70 (br.s., 1 H), 4.71 (br.s., 1 H), 3.78-3.43 (m, 2 H), 2.58-2.39 (m, 4 H), 2.10-2.04 (m, 1 H), 1.84-1.79 (m, 1H), 1.76- 1.62 (m, 2H), 1.54-1.40 (m, 1H), 1.14-0.99 (m, 1H), 0.56-0.02 (m, 1H), −.027-−0.83 (m, 1H). ESI+ m/z 468 [M + H]+
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
4b (D58) 27 15
Figure US09174977-20151103-C00114
1H NMR (CDCl3) δ ppm = 8.67 (br. s., 2 H), 8.36-8.34 (m, 2 H), 7.57-7.49 (m, 2 H), 7.45-7.31 (m, 1H), 7.20-7.17 (m, 1 H,), 6.69- 6.67 (m, 1 H), 4.64 (m 1 H), 3.75-3.59 (m, 1 H), 3.56-3.45 (m, 1 H), 2.49 (m, 1 H), 2.12-2.03 (m, 1H), 1.85-1.65 (m, 3H), 1.61-1.40 (m, 1H), 1.18-1.02 (m, 1H), 0.63-0.01 (m, 1H), −.007- −0.87 (m, 1H). ESI+ m/z 488 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
4c (D56) 27 11
Figure US09174977-20151103-C00115
1H NMR (CDCl3) δ ppm = 8.73 (br. s., 2 H), 8.53 (d, J = 8 Hz,1 H), 8.35 (s, 1 H),7.82 (dd, J = 8 Hz, 2 Hz, 1H), 7.72 (m, 1 H,), 7.56 (dd, J = 8 Hz, 2 Hz, 1 H), 7.27 (m, 1 H, under solvent peak), 6.67 (d, J = 8 Hz, 1 H), 4.61 (m, 1 H), 3.73-3.62 (m, 1H), 3.55-3.49 (m, 1H), 2.53-2.36 (m, 1H), 2.14-2.09 (m, 1H), 1.87-1.66 (m, 3H), 1.60-1.42 (m, 1H), 1.21-1.08 (m, 1H), 0.60-−0.04 ((m, 1H), −0.10-−1.01 (m, 1H). ESI+ m/z 479 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
4e (D60) 29 12
Figure US09174977-20151103-C00116
1H NMR (CDCl3) δ ppm = 8.68 (br. s., 2 H), 8.36-8.34 (m, 1 H), 8.04-8.05 (m, 1H), 7.52-7.49 (m, 1 H), 7.44-7.31 (m, 2H), 7.20- 7.18 (m, 1 H,), 6.66-6.64 (m, 1 H), 4.64 (m, 1 H), 3.71-3.56 (m, 1 H), 3.42-3.36 (m, 1 H), 2.56-2.41 (m, 1H), 2.13-2.03 (m, 1H), 1.86- 1.64 (m, 3H), 1.54-1.40 (m, 1H), 1.22-1.02 (m, 1H), 0.61-0.02 (m, 1H), −.01-−1.05 (m, 1H). ESI+ m/z 454 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
4f (D59) 29 47
Figure US09174977-20151103-C00117
1H NMR (CDCl3) δ ppm = 8.62 (br. s., 2 H), 8.28-8.23 (m, 2 H), 7.35-7.33 (m, 1H), 7.25-7.07 (m, 2 H), 6.75-6.71 (m, 2H), 4.76 (m, 1 H), 3.67-3.50 (m, 2 H), 2.60-2.45 (m, 4 H), 2.11-2.02 (m, 1H), 1.85-1.66 (m, 2H), 1.57-1.44 (m, 2H, under water peak), 1.13-0.98 (m, 1H), 0.57-0.01 (m, 1H), −0.24-−0.95 (m, 1H). ESI+ m/z 468 [M + H]+
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
4g (D62) 29 30
Figure US09174977-20151103-C00118
1H NMR (CDCl3) δ ppm = 8.66 (br. s., 2 H), 8.27-8.25 (m, 1 H), 8.05-8.02 (m, 1H), 7.37-7.33 (m, 2H), 7.25-7.13 (m, 2 H), 6.75- 6.66 (m, 1H), 4.69 (m, 1 H), 3.67-3.52 (m, 1 H), 3.41-3.35 (m, 1H), 2.52-2.45 (m, 4 H), 2.10-2.02 (m, 1H), 1.84-1.80 (m, 1H), 1.75-1.55 (m, 3H), 1.12-0.98 (m, 1H), 0.66-0.01 (m, 1H), −0.21-−0.82 (m, 1H). ESI+ m/z 434 [M + H]+
((1S,3S,6R)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-
(pyrimidin-2-yl)phenyl)methanone
4h (D63) 29 44
Figure US09174977-20151103-C00119
1H NMR (CDCl3) δ ppm = 8.81 (m, 2 H), 8.50 (d, J = 4 Hz, 1 H), 8.36 (d, J = 8 Hz, 1H), 7.50 (dd, J = 8 Hz, 4 Hz, 1H), 7.23 (m, 1H), 6.83 (d, J = 4 Hz, 1 H), 6.18 (m, 1 H), 4.49 (m, 1H), 3.84-3.63 (m, 2 H), 2.49 (m, 1H), 2.10-1.99 (m, 1H), 1.79-1.51 (m, 3H, under solvent peak), 1.10 (m, 1H), 0.66-0.01 (m, 1H), −0.19-−0.72 (m, 1H). ESI+ m/z 489 [M + H]+
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
4i (D64) 29 34
Figure US09174977-20151103-C00120
1H NMR (CDCl3) δ ppm = 8.81 (m, 2 H), 8.50 (d, J = 4 Hz, 1 H), 8.28 (d, J = 8 Hz, 1H), 7.33 (d, J = 8 Hz, 1H), 7.18 (m, 1 H), 6.83 (d, J = 4 Hz, 1 H), 6.28 (m, 1 H), 4.53 (m, 1H), 3.82-3.67 (m, 2 H), 2.53-2.49 (m, 4H), 2.08-1.97 (m, 1H), 1.79-1.52 (m, 3H, under solvent peak), 1.04 (m, 1H), 0.60-0.04 (m, 1H), −0.09-−0.80 (m, 1H). ESI+ m/z 469 [M + H]+
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-
azabicyclo[4.1.0]heptan-2-yl)methanone
Example 5 Preparation of Compound 5a (2-methyl-5-phenylthiazol-4-yl)((1S,3S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone
Figure US09174977-20151103-C00121
D65 (12 mg, 0.06 mmol), N-methyl-morpholine (22 μL, 0.20 mmol) and 2-chloro-4,6-dimethoxy-1,3,5-triazine (12 mg, 0.06 mmol) dissolved in dry 1,4-dioxane (0.5 mL) were stirred at 25° C. for 0.5 hour, then D52 (18 mg, 0.06 mmol) dissolved in 1,4-dioxane (0.5 mL) was added. The reaction mixture was heated overnight at 60° C., cooled to RT and purified by silica gel column chromatography (Cyclohexane to AcOEt). The residue was dissolved in DCM, washed with an aqueous saturated solution of NaHCO3 and concentrated to give the title compound. Yield (21 mg, 77%)
1H NMR (CDCl3) δ ppm 8.35 (m, 1H), 7.56 (m, 1H), 7.46 (m, 2H), 7.30-7.32 (m, 1H), 7.19 (m, 2H), 6.62 (m, 1H), 6.11 (m, 1H), 4.62 (m, 1H), 3.56-3.62 (m, 1H), 3.35-3.43 (m, 1H), 2.77 (s, 3H), 2.66 (m, 1H), 2.01 (m, 1H), 1.67-1.76 (m, 2H), 1.43-1.51 (m, 1H), 1.07 (m, 1H), 0.34 (m, 1H), −0.21 (m, 1H).
ESI+ m/z 472 [M+H]+
BIOLOGICAL SECTION
In a typical experiment, the antagonistic activity against human OX1 and OX2 receptors is determined by using CHO e HEK-293 cells transfected with human recombinant OX1 and OX2 receptors respectively, seeded at density of 2 and 3×104 cells/well respectively in a 96 fluorimetry well plate. Thus the plate was loaded with the calcium dye (Fluo-4NW/probenecid in HBSS, Hepes 20 mM, pH 7,4; Invitrogen) at 37° C. for 60 min. Afterward the temperature was equilibrated at 22° C. for 15 min and the [Ca2+]i measured directly on the plate by using a fluorescent plate reader (CellLux Perkin Elmer).
Invention compounds were dissolved in DMSO, diluted in HBSS (DMSO, 0.3% final) and added to the wells. After 5 min CHO cells were activated with orexin-A, 3 nM while HEK-293 cells were activated with orexin-B, 10 nM.
The compounds, dissolved in DMSO and diluted in the medium (DMSO, 0.3% final), have been analysed in the 1 nM-1 μM concentration range (every concentration in duplicate). The antagonistic activity has been expressed as pKb (co-logarithm of the apparent dissociation constant calculated by using the modified Cheng Prusoff equation).
The results are expressed as percent of control specific antagonist response ((measured specific response/control specific agonist response)×100) obtained in the presence of the test compounds.
The IC50 values (concentration causing a half-maximal inhibition of the control specific agonist response) were determinated by non-linear regression analysis of the concentration curves generated with mean replicate values using hill equation curve fitting. The IC50 values are obtained by the arithmetical mean of at least two experiments.
Compounds of the following example tested according to this example gave pKbs as follows:
Compound pKb OX1 pKb OX2
1a 8.8 8.4
1b 8.3 8.0
1c 9.1 7.9
1d 8.9 8.4
1e 8.6 7.9
1f 9.1 7.8
1g 9.0 8.3
1h 8.1 8.0
1i 8.8 7.7
1j 8.3 7.7
1k 8.6 8.3
2a 8.6 8.6
2b 8.9 7.8
2c 8.9 8.2
3a 8.9 7.2
3b 7.1 5.0
3c 7.9 7.2
3d 8.9 7.7
3e 9.0 7.8
3f 9.0 7.3
3g 8.8 7.9
4a 8.6 7.4
4b 8.3 6.7
4c 8.2 7.1
4d 8.1 7.3
4e 8.4 5.0
4f 8.2 7.4
4g 8.8 5.0
4h 7.5 5.0
4i 7.6 5.0
5a 8.6 7.3

Claims (7)

The invention claimed is:
1. A compound of formula (II) or a pharmaceutically acceptable salt thereof:
Figure US09174977-20151103-C00122
wherein
Q is 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
R and R1 are independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
m is 0, 1, 2, 3, or 4; and
n is 1, 2, or 3.
2. A compound of formula (III) or a pharmaceutically acceptable salt thereof:
Figure US09174977-20151103-C00123
wherein
Q is 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
R and R1 are independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
m is 0, 1, 2, 3, or 4; and
P is 0 or 1.
3. A compound of formula (V) or a pharmaceutically acceptable salt thereof:
Figure US09174977-20151103-C00124
wherein
Q is 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
R and R1 are independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
m is 0, 1, 2, 3, or 4;
n is 1, 2, or 3.
4. A compound of formula (VI) or a pharmaceutically acceptable salt thereof:
Figure US09174977-20151103-C00125
wherein
Q is 5-6 membered heteroaryl group, which may be substituted by one or more substituents independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
R and R1 are independently selected from: C1-C4 alkyl, halogen, halo C1-C4 alkyl, C1-C4 alkoxy, or CN;
m is 0, 1, 2, 3, or 4; and
p is 0 or 1.
5. A compound, wherein the compound is:
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-4(5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
((1R,3S,6S)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-(pyrimidin-2-yl)phenyl)methanone;
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,65)-3-4(5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,65)-3-4(5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,65)-3-4(5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1R,3S,65)-3-4(4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
4-(pyrimidin-2-yl)-3-((1R,3S,65)-3-4(4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
3-((1R,3S,65)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)-4-(pyrimidin-2-yl)benzonitrile;
4-(pyrimidin-2-yl)-3-((1R,3S,6S)-3-4(4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1R,3S,6S)-3-4(4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(2-methyl-5-phenylthiazol-4-yl)((1R,3S,65)-3-4(5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
((1R,3S,65)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(2-methyl-5-phenylthiazol-4-yl)methanone;
(2-methyl-5-phenylthiazol-4-yl)((1R,3S,65)-3-((4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3 S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-4(5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
4-(pyrimidin-2-yl)-3-((1S,3 S,6R)-3-(((5-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptane-2-carbonyl)benzonitrile;
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-4(4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-4(5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-4(4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
((1S,3S,6R)-3-(((5-chloropyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)(5-methyl-2-(pyrimidin-2-yl)phenyl)methanone;
(5-chloro-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
(5-methyl-2-(pyrimidin-2-yl)phenyl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyrimidin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone; or
(2-methyl-5-phenylthiazol-4-yl)((1S,3S,6R)-3-(((4-(trifluoromethyl)pyridin-2-yl)amino)methyl)-2-azabicyclo[4.1.0]heptan-2-yl)methanone;
or a pharmaceutically acceptable salt thereof.
6. A method for treating obesity, sleep disorders, compulsive disorders, substance abuse, or schizophrenia comprising administering to a subject in need thereof an effective amount of a compound as in any one of claims 1, 2, 3, 4, and 5 or a pharmaceutically acceptable salt thereof.
7. A pharmaceutical composition comprising a compound as in any one of claims 1, 2, 3, 4, and 5 or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
US14/386,109 2012-03-19 2013-03-18 2-azabicyclo[4.1.0]heptane derivatives as orexin receptor antagonists for the treatment of certain disorders Expired - Fee Related US9174977B2 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
ITMI2012A0424 2012-03-19
IT000424A ITMI20120424A1 (en) 2012-03-19 2012-03-19 CHEMICAL COMPOUNDS
ITMI2012A000424 2012-03-19
PCT/EP2013/055548 WO2013139730A1 (en) 2012-03-19 2013-03-18 Chemical compounds

Publications (2)

Publication Number Publication Date
US20150065523A1 US20150065523A1 (en) 2015-03-05
US9174977B2 true US9174977B2 (en) 2015-11-03

Family

ID=46028058

Family Applications (1)

Application Number Title Priority Date Filing Date
US14/386,109 Expired - Fee Related US9174977B2 (en) 2012-03-19 2013-03-18 2-azabicyclo[4.1.0]heptane derivatives as orexin receptor antagonists for the treatment of certain disorders

Country Status (9)

Country Link
US (1) US9174977B2 (en)
EP (1) EP2827870B1 (en)
JP (1) JP6174112B2 (en)
AR (1) AR090350A1 (en)
CA (1) CA2866410A1 (en)
ES (1) ES2587952T3 (en)
IT (1) ITMI20120424A1 (en)
TW (1) TW201341379A (en)
WO (1) WO2013139730A1 (en)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9440982B2 (en) 2012-02-07 2016-09-13 Eolas Therapeutics, Inc. Substituted prolines/piperidines as orexin receptor antagonists
ES2672732T3 (en) 2012-02-07 2018-06-15 Eolas Therapeutics Inc. Prolines / substituted piperidines as orexin receptor antagonists
UY36272A (en) 2014-08-13 2016-02-29 Eolas Therapeutics Inc DIFLUOROPIRROLIDINS AS MODULATORS OF OREXIN RECEPTORS
KR20180107232A (en) 2016-02-12 2018-10-01 아스트라제네카 아베 The halo-substituted piperidine as an orexin receptor modulator
US11660293B2 (en) 2017-09-01 2023-05-30 Chronos Therapeutics Limited Substituted 2-azabicyclo[3.1.1]heptane and 2-azabicyclo[3.2.1]octane derivatives as orexin receptor antagonists
GB2558975B (en) * 2017-09-01 2019-01-23 Chronos Therapeutics Ltd Substituted 2-azabicyclo[3.1.1]heptane and 2-azabicyclo[3.2.1]octane derivatives as orexin receptor antagonists
WO2019081939A1 (en) * 2017-10-25 2019-05-02 Chronos Therapeutics Limited 2-azabicyclo[3.1.1] derivatives as antagonists of the orexin-1 and orexin-2 receptors
JP7365535B1 (en) 2022-03-18 2023-10-19 Nok株式会社 valve device

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3282927A (en) 1964-05-21 1966-11-01 Bristol Myers Co 5-phenyl-4-thiazolylpenicillins
WO2003051368A1 (en) 2001-12-19 2003-06-26 Smithkline Beecham Plc N-aroyl cyclic amine derivatives as orexin receptor antagonists
WO2006127550A1 (en) 2005-05-23 2006-11-30 Merck & Co., Inc. Proline bis-amide orexin receptor antagonists
WO2008038251A2 (en) 2006-09-29 2008-04-03 Actelion Pharmaceuticals Ltd 3-aza-bicyclo[3.1.0]hexane derivatives
WO2008081399A2 (en) 2006-12-28 2008-07-10 Actelion Pharmaceuticals Ltd 2-aza-bicyclo[3.1.0]hexane derivatives as orexin receptor antagonists
WO2008101665A1 (en) 2007-02-20 2008-08-28 Novartis Ag Macrocyclic compounds as hcv ns3 protease inhibitors
WO2008139416A1 (en) 2007-05-14 2008-11-20 Actelion Pharmaceuticals Ltd 2-cyclopropyl-thiazole derivatives
WO2008147518A1 (en) 2007-05-23 2008-12-04 Merck & Co., Inc. Pyridyl piperidine orexin receptor antagonists
WO2009016564A2 (en) 2007-07-27 2009-02-05 Actelion Pharmaceuticals Ltd 2-aza-bicyclo[3.3.0]octane derivatives
WO2010048016A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,3-disubstituted piperidine orexin receptor antagonists
WO2010051238A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Pyridazine carboxamide orexin receptor antagonists
WO2010060470A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
WO2010060471A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
WO2010063662A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0] hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
WO2010063663A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0]hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
WO2010122151A1 (en) 2009-04-24 2010-10-28 Glaxo Group Limited 3 -azabicyclo [4.1.0] heptanes used as orexin antagonists
WO2010132601A1 (en) 2009-05-13 2010-11-18 Gilead Sciences, Inc. Antiviral compounds
EP2275421A1 (en) 2009-07-15 2011-01-19 Rottapharm S.p.A. Spiro amino compounds suitable for the treatment of inter alia sleep disorders and drug addiction
WO2011076747A1 (en) 2009-12-21 2011-06-30 Novartis Ag Diaza-spiro[5.5]undecanes as orexin receptor antagonists
WO2012104338A1 (en) 2011-02-02 2012-08-09 Rottapharm S.P.A. Spiro aminic compounds with nk1 antagonist activity
WO2013092893A1 (en) 2011-12-21 2013-06-27 Rottapharm Spa Chemical compounds

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0225884D0 (en) * 2002-11-06 2002-12-11 Glaxo Group Ltd Novel compounds
US20100099981A1 (en) 2008-10-21 2010-04-22 Fishel Robert S Trans-Septal Catheterization Device And Method

Patent Citations (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3282927A (en) 1964-05-21 1966-11-01 Bristol Myers Co 5-phenyl-4-thiazolylpenicillins
WO2003051368A1 (en) 2001-12-19 2003-06-26 Smithkline Beecham Plc N-aroyl cyclic amine derivatives as orexin receptor antagonists
WO2006127550A1 (en) 2005-05-23 2006-11-30 Merck & Co., Inc. Proline bis-amide orexin receptor antagonists
WO2008038251A2 (en) 2006-09-29 2008-04-03 Actelion Pharmaceuticals Ltd 3-aza-bicyclo[3.1.0]hexane derivatives
WO2008081399A2 (en) 2006-12-28 2008-07-10 Actelion Pharmaceuticals Ltd 2-aza-bicyclo[3.1.0]hexane derivatives as orexin receptor antagonists
WO2008101665A1 (en) 2007-02-20 2008-08-28 Novartis Ag Macrocyclic compounds as hcv ns3 protease inhibitors
WO2008139416A1 (en) 2007-05-14 2008-11-20 Actelion Pharmaceuticals Ltd 2-cyclopropyl-thiazole derivatives
WO2008147518A1 (en) 2007-05-23 2008-12-04 Merck & Co., Inc. Pyridyl piperidine orexin receptor antagonists
WO2009016564A2 (en) 2007-07-27 2009-02-05 Actelion Pharmaceuticals Ltd 2-aza-bicyclo[3.3.0]octane derivatives
WO2010048016A1 (en) 2008-10-21 2010-04-29 Merck Sharp & Dohme Corp. 2,3-disubstituted piperidine orexin receptor antagonists
WO2010051238A1 (en) 2008-10-30 2010-05-06 Merck Sharp & Dohme Corp. Pyridazine carboxamide orexin receptor antagonists
WO2010060471A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
WO2010060470A1 (en) 2008-11-26 2010-06-03 Glaxo Group Limited Piperidine derivatives useful as orexin receptor antagonists
US8133908B2 (en) * 2008-12-02 2012-03-13 Glaxo Group Limited Heteroaryl derivatives of N-{[(1S,4S,6S)-3-(2-pyridinylcarbonyl)-3-azabicyclo[4.1.0]hept-4-yl]methyl}-2-amine
WO2010063662A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0] hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
WO2010063663A1 (en) 2008-12-02 2010-06-10 Glaxo Group Limited N-{[(ir,4s,6r-3-(2-pyridinylcarbonyl)-3-azabicyclo [4.1.0]hept-4-yl] methyl}-2-heteroarylamine derivatives and uses thereof
WO2010122151A1 (en) 2009-04-24 2010-10-28 Glaxo Group Limited 3 -azabicyclo [4.1.0] heptanes used as orexin antagonists
US20120040991A1 (en) * 2009-04-24 2012-02-16 Glaxo Group Limited 3-azabicyclo [4.1.0] heptanes used as orexin antagonists
WO2010132601A1 (en) 2009-05-13 2010-11-18 Gilead Sciences, Inc. Antiviral compounds
EP2275421A1 (en) 2009-07-15 2011-01-19 Rottapharm S.p.A. Spiro amino compounds suitable for the treatment of inter alia sleep disorders and drug addiction
WO2011006960A1 (en) 2009-07-15 2011-01-20 Rottapharm S.P.A. Spiro amino compounds suitable for the treatment of inter alia sleep disorders and drug addiction
US8859608B2 (en) 2009-07-15 2014-10-14 Rottapharm Biotech S.R.L. Spiro amino compounds suitable for the treatment of inter alia sleep disorders and drug addiction
WO2011076747A1 (en) 2009-12-21 2011-06-30 Novartis Ag Diaza-spiro[5.5]undecanes as orexin receptor antagonists
WO2012104338A1 (en) 2011-02-02 2012-08-09 Rottapharm S.P.A. Spiro aminic compounds with nk1 antagonist activity
WO2013092893A1 (en) 2011-12-21 2013-06-27 Rottapharm Spa Chemical compounds
US20140357653A1 (en) 2011-12-21 2014-12-04 Rottapharm Biotech S.R.I. Chemical compounds

Non-Patent Citations (38)

* Cited by examiner, † Cited by third party
Title
Aston-Jones et al., 2009, "Role of lateral hypothalamic orexin neurons in reward processing and addiction," Neuropharmacology, 56:112-121.
Brisbare-Roch et al., 2009, "Promotion of sleep by targeting the orexin system in rats, dogs and humans," Nature Medicine (Advance Online Publication), 1-6.
Chemelli et al., 1999, "Narcolepsy in orexin Knockout Mice: Molecular Genetics of Sleep Regulation," Cell, 98:437-451.
Chen et al., 2000, "Pressor effects of orexins injected intracisternally and to rostral ventrolateral medulla of anesthetized rats," Am. J. Physiol. Reg. Integrative Comp. Physiol., 278:R692-R697.
Dayas et al., 2008, "Stimuli Linked to Ethanol Availability Activate Hypothalamic CART and Orexin Neurons in a Reinstatement Model of Relapse," Biol. Psychiatry, 63:152-157.
Dugovic et al., 2009, "Blockade of Orexin-1 Receptors Attenuates Orexin-2 Receptor Antagonism-Induced Sleep Promotion in the Rat," The Journal of Pharmacology and Experimental Therapeutics, 330:142-151.
Fargeas et al., 2003, "Nitration of Heteroaryltrimethyltins by Tetranitromethane and Dinitrogen Tetroxide: Mechanistic Aspects, Scope and Limitations," Eur. J. Org. Chem., 1711-1721.
Fleisher et al., 1996, "Imporved oral drug delivery: solubility limitations overcome by the use of prodrugs," Advanced Drug Delivery Reviews, 19:115-130.
Georgescu et al., 2003, "Involvement of the Lateral Hypothalamic Peptide Orexin in Morphine Dependence and Withdrawal," J. Neurosci., 23(8):3106-3111.
Hamlin et al., 2007, "The Neural Correlates and Role of D1 Dopamine Receptors in Renewal of Extinguished Alcohol-Seeking," Neuroscience 146:525-536.
Hara et al., 2001, "Genetic Ablation of Orexin Neurons in Mice Results in Narcolepsy, Hypophagia, and Obesity," Neuron, 30:345-354.
International Search Report mailed Mar. 5, 2013 for International Patent Application No. PCT/EP2012/076447, filed Dec. 20, 2012.
Kane et al., 2000, "Nicotine Up-Regulates Expression of Orexin and Its Receptors in Rat Brain," Endocrinology, 141:3623-3629.
Kane et al., 2001, "Hypothalamic orexin-A binding sites are downregulated by chronic nicotine treatment in the rat," Neuroscience Letters 298:1-4.
Kang et al., 2009, "Amyloid-beta Dynamics Are Regulated by Orexin and the Sleep-Wake Cycle," Science, 326:1005-1007.
Kang et al., 2009, "Amyloid-β Dynamics Are Regulated by Orexin and the Sleep-Wake Cycle," Science, 326:1005-1007.
Kirchgessner et al., 1999, "Orexin Synthesis and Response in the Gut," Neuron, 24:941-951.
Lawrence et al., 2006, "The orexin system regulates alcohol-seeking in rats," British Journal of Pharmacology, 148:752-759.
Lin et al., 1999, "The Sleep Disorder Canine Narcolepsy is Caused by a Mutation in the Hypocretin (Orexin) Receptor 2 Gene," Cell, 98:365-376.
Malherbe et al., 2009, "Biochemical and Electrophysiological Characterization of Almorexant, a Dual Orexin 1 Receptor (OX1)/Orexin 2 Receptor (OX2) Antagonist: Comparison with Selective OX1 and OX2 Antagonists," Molecular Pharmacology, 76:618-631.
Mignot et al., 2001, "Complex HLA-DR and -DQ Interactions Confer Risk of Narcolepsy-Cataplexy in Three Ethnic Groups," Am. J. Hum. Genet., 68:686-699.
Minot et al., 2001, "Narcolepsy and the HLA System," N. Engl. J. Med., 344:686-693 at p. 692.
Nakamura et al., 2000, "Orexin-induced hyperlocomotion and stereotypy are mediated by the dopaminergic system," Brain Research 873:181-187.
Peyron et al., 1998, "Neurons Containing Hypocretin (Orexin) Project to Multiple Neuronal Systems," Journal of Neuroscience, 18(23):9996-10015.
Peyron et al., 2000, "A mutation in a case of early onset narcolepsy and a generalized absence of hypocretin peptides in human narcoleptic brains," Nature Medicine, 6:991-997.
Piper et al., 2000, "The novel brain neuropeptide, orexin-A, modulates the sleep-wake cycle of rats," European Journal of Neuroscience, 12:726-730.
Richards et al., 2008, "Inhibition of orexin-1/hypocretin-1 receptors inhibits yohimbine-induced reinstatement of ethanol and sucrose seeking in long-evans rats," Psychopharmacology, 199:109-117.
Sakurai et al., 1998, "Orexins and Orexin Receptors: A Family of Hypothalamic Neuropeptides and G Protein-Coupled Receptors That Regulate Feeding Behavior," Cell, 92:573-585.
Sakurai, 1999, "Orexins and orexin receptors: implication in feeding behavior," Regulatory Peptides, 85:25-30.
Sakurai, 2007, "The neural circuit of orexin (hypocretin): maintaining sleep and wakefulness," Nature Reviews Neuroscience, 8:171-181.
Samson et al., 1999, "Cardiovascular regulatory actions of the hypocretins in brain," Brain Research, 831:248-253.
Schneider et al., 2007, "Orexigenic Peptides and Alcohol Intake: Differential Effects of Orexin, Galanin, and Ghrelin," Alcoholism: Clinical and Experimental Research, 31:1-8.
Shirasaka et al., 1999, "Sympathetic and cardiovascular actions of orexins in conscious rats," Am. J. Physiol. 277 (Regulatory Integrative Comp. Physiol. 46): R1780-R1785.
Takahashi et al., 1999, "Stimulation of Gastric Acid Secretion by Centrally Administered Orexin-A in Conscious Rats," Biochemical and Biophysical Research Communications, 254:623-627.
Van Den Pol, 1999, "Hypothalamic Hypocretin (Orexin): Robust Innervation of the Spinal Cord," Journal of Neuroscience, 19:3171-3182.
Winrow et al., 2010, "Orexin receptor antagonism prevents transcriptional and behavioral plasticity resulting from stimulant exposure," Neuropharmacology, 58:185-194.
Written Opinion of the International Searching Authority mailed May 3, 2013 for International Patent Application No. PCT/EP2012/076447, filed Dec. 20, 2012.
Yamanaka et al., 2002, "Orexins Activate Histaminergic Neurons via the Orexin 2 Receptor," Biochemical and Biophysical Research Communications, 290:1237-1245.

Also Published As

Publication number Publication date
WO2013139730A1 (en) 2013-09-26
EP2827870B1 (en) 2016-05-25
JP2015510909A (en) 2015-04-13
TW201341379A (en) 2013-10-16
ES2587952T3 (en) 2016-10-27
US20150065523A1 (en) 2015-03-05
CA2866410A1 (en) 2013-09-26
EP2827870A1 (en) 2015-01-28
ITMI20120424A1 (en) 2013-09-20
AR090350A1 (en) 2014-11-05
JP6174112B2 (en) 2017-08-02

Similar Documents

Publication Publication Date Title
US9174977B2 (en) 2-azabicyclo[4.1.0]heptane derivatives as orexin receptor antagonists for the treatment of certain disorders
JP6204484B2 (en) Heteroaryl-substituted pyridyl compounds useful as kinase modulators
JP5281395B2 (en) Cyclohexanesulfonyl derivatives as GLYT1 inhibitors for the treatment of schizophrenia
US9062033B2 (en) Chemical compounds
US10533000B2 (en) Metabotrophic glutamate receptor 5 modulators and methods of use thereof
JP2017501236A (en) Pyrrolidinyl sulfone derivatives and their use as RORγ regulators
WO1999050264A1 (en) Quinazoline derivatives
JP2023502441A (en) SUBSTITUTED MACROCYCLES AND RELATED THERAPEUTIC METHODS
KR20070026382A (en) Acetylinic piperazine compounds and their use as metabotropic glutamate receptor antagonists
JP2017514856A (en) Pyrrolidine GPR40 modulator for the treatment of diseases such as diabetes
JP2024501641A (en) Substituted Macrocycles and Related Treatment Methods
JP2024517678A (en) Modulators of sortilin activity
JP6488320B2 (en) Pyrrolidine GPR40 modulator for the treatment of diseases such as diabetes
CN102933575B (en) Yclohexyl-azetidinyl antagonists of CCR2
JP7386797B2 (en) Substituted cyclohexyl compounds as NOP inhibitors
US11970493B2 (en) Autotaxin inhibitor compounds
US20090176836A1 (en) Cyclic Ketal Beta-Secretase Inhibitors for the Treatment of Alzheimer's Disease
AU2022267220A1 (en) Substituted amide macrocyclic compounds with orexin-2 receptor agonist activity
EP4347604A1 (en) Substituted carbamate macrocyclic compounds and related methods of treatment
EP3009421A1 (en) Glycine transporter inhibitor
WO2023249872A1 (en) Substituted fused bicyclic compounds and related methods of treatment
WO2022251302A1 (en) Substituted fused bicyclic macrocyclic compounds and related methods of treatment
TW202346289A (en) Compounds as glp-1r agonists
WO2003078441A1 (en) Aminomethyl-substituted thiazolobenzimidazole derivative
TW202400606A (en) Medium- or macro-cyclic benzyl-substituted heterocycle derivatives and related uses

Legal Events

Date Code Title Description
AS Assignment

Owner name: ROTTAPHARM BIOTECH S.R.L., ITALY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:STASI, LUIGI PIERO;ROVATI, LUCIO CLAUDIO;REEL/FRAME:035289/0909

Effective date: 20141112

STCF Information on status: patent grant

Free format text: PATENTED CASE

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Expired due to failure to pay maintenance fee

Effective date: 20191103